1
|
Balough JL, Dipali SS, Velez K, Kumar TR, Duncan FE. Hallmarks of female reproductive aging in physiologic aging mice. NATURE AGING 2024; 4:1711-1730. [PMID: 39672896 DOI: 10.1038/s43587-024-00769-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/28/2024] [Indexed: 12/15/2024]
Abstract
The female reproductive axis is one of the first organ systems to age, which has consequences for fertility and overall health. Here, we provide a comprehensive overview of the biological process of female reproductive aging across reproductive organs, tissues and cells based on research with widely used physiologic aging mouse models, and describe the mechanisms that underpin these phenotypes. Overall, aging is associated with dysregulation of the hypothalamic-pituitary-ovarian axis, perturbations of the ovarian stroma, reduced egg quantity and quality, and altered uterine morphology and function that contributes to reduced capacity for fertilization and impaired embryo development. Ultimately, these age-related phenotypes contribute to altered pregnancy outcomes and adverse consequences in offspring. Conserved mechanisms of aging, as well as those unique to the reproductive system, underlie these phenotypes. The knowledge of such mechanisms will lead to development of therapeutics to extend female reproductive longevity and support endocrine function and overall health.
Collapse
Affiliation(s)
- Julia L Balough
- Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, USA
| | - Shweta S Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Karen Velez
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - T Rajendra Kumar
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Francesca E Duncan
- Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, USA.
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
2
|
Sun F, Ali NN, Londoño-Vásquez D, Simintiras CA, Qiao H, Ortega MS, Agca Y, Takahashi M, Rivera RM, Kelleher AM, Sutovsky P, Patterson AL, Balboula AZ. Increased DNA damage in full-grown oocytes is correlated with diminished autophagy activation. Nat Commun 2024; 15:9463. [PMID: 39487138 PMCID: PMC11530536 DOI: 10.1038/s41467-024-53559-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/14/2024] [Indexed: 11/04/2024] Open
Abstract
Unlike mild DNA damage exposure, DNA damage repair (DDR) is reported to be ineffective in full-grown mammalian oocytes exposed to moderate or severe DNA damage. The underlying mechanisms of this weakened DDR are unknown. Here, we show that moderate DNA damage in full-grown oocytes leads to aneuploidy. Our data reveal that DNA-damaged oocytes have an altered, closed, chromatin state, and suggest that the failure to repair damaged DNA could be due to the inability of DDR proteins to access damaged loci. Our data also demonstrate that, unlike somatic cells, mouse and porcine oocytes fail to activate autophagy in response to DNA double-strand break-inducing treatment, which we suggest may be the cause of the altered chromatin conformation and inefficient DDR. Importantly, autophagy activity is further reduced in maternally aged oocytes (which harbor severe DNA damage), and its induction is correlated with reduced DNA damage in maternally aged oocytes. Our findings provide evidence that reduced autophagy activation contributes to weakened DDR in oocytes, especially in those from aged females, offering new possibilities to improve assisted reproductive therapy in women with compromised oocyte quality.
Collapse
Affiliation(s)
- Fei Sun
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Nourhan Nashat Ali
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Physiology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | | | - Constantine A Simintiras
- School of Animal Sciences, Agricultural Center, Louisiana State University, Baton Rouge, LA, USA
| | - Huanyu Qiao
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - M Sofia Ortega
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuksel Agca
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Masashi Takahashi
- Research Faculty of Agriculture, Hokkaido University, Hokkaido, Japan
| | - Rocío M Rivera
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Andrew M Kelleher
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Peter Sutovsky
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Amanda L Patterson
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Ahmed Z Balboula
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
3
|
Zhu Y, Kratka CR, Pea J, Lee HC, Kratka CE, Xu J, Marin D, Treff NR, Duncan FE. The severity of meiotic aneuploidy is associated with altered morphokinetic variables of mouse oocyte maturation. Hum Reprod Open 2024; 2024:hoae023. [PMID: 38764910 PMCID: PMC11099657 DOI: 10.1093/hropen/hoae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/04/2024] [Indexed: 05/21/2024] Open
Abstract
STUDY QUESTION Is there an association between morphokinetic variables of meiotic maturation and the severity of aneuploidy following in vitro maturation (IVM) in the mouse? SUMMARY ANSWER The severity of meiotic aneuploidy correlates with an extended time to first polar body extrusion (tPB1) and duration of meiosis I (dMI). WHAT IS KNOWN ALREADY Morphokinetic variables measured using time-lapse technology allow for the non-invasive evaluation of preimplantation embryo development within clinical assisted reproductive technology (ART). We recently applied this technology to monitor meiotic progression during IVM of mouse gametes. Whether there is a relationship between morphokinetic variables of meiotic progression and aneuploidy in the resulting egg has not been systematically examined at the resolution of specific chromosomes. Next-generation sequencing (NGS) is a robust clinical tool for determining aneuploidy status and has been reverse-translated in mouse blastocysts and oocytes. Therefore, we harnessed the technologies of time-lapse imaging and NGS to determine the relationship between the morphokinetics of meiotic progression and egg aneuploidy. STUDY DESIGN SIZE DURATION Cumulus-oocyte complexes were collected from large antral follicles from hyperstimulated CD-1 mice. Cumulus cells were removed, and spontaneous IVM was performed in the absence or presence of two doses of Nocodazole (25 or 50 nM) to induce a spectrum of spindle abnormalities and chromosome segregation errors during oocyte meiosis. Comprehensive chromosome screening was then performed in the resulting eggs, and morphokinetic variables and ploidy status were compared across experimental groups (control, n = 11; 25 nM Nocodazole, n = 13; 50 nM Nocodazole, n = 23). PARTICIPANTS/MATERIALS SETTING METHODS We monitored IVM in mouse oocytes using time-lapse microscopy for 16 h, and time to germinal vesicle breakdown (tGVBD), tPB1, and dMI were analyzed. Following IVM, comprehensive chromosome screening was performed on the eggs and their matched first polar bodies via adaptation of an NGS-based preimplantation genetic testing for aneuploidy (PGT-A) assay. Bioinformatics analysis was performed to align reads to the mouse genome and determine copy number-based predictions of aneuploidy. The concordance of each polar body-egg pair (reciprocal errors) was used to validate the results. Ploidy status was categorized as euploid, 1-3 chromosomal segregation errors, or ≥4 chromosomal segregation errors. Additionally, aneuploidy due to premature separation of sister chromatids (PSSC) versus non-disjunction (NDJ) was distinguished. MAIN RESULTS AND THE ROLE OF CHANCE We applied and validated state-of-the-art NGS technology to screen aneuploidy in individual mouse eggs and matched polar bodies at the chromosome-specific level. By performing IVM in the presence of different doses of Nocodazole, we induced a range of aneuploidy. No aneuploidy was observed in the absence of Nocodazole (0/11), whereas IVM in the presence of 25 and 50 nM Nocodazole resulted in an aneuploidy incidence of 7.69% (1/13) and 82.61% (19/23), respectively. Of the aneuploid eggs, 5% (1/20) was due to PSSC, 65% (13/20) to NDJ, and the remainder to a combination of both. There was no relationship between ploidy status and tGVBD, but tPB1 and the dMI were both significantly prolonged in eggs with reciprocal aneuploidy events compared to the euploid eggs, and this scaled with the severity of aneuploidy. Eggs with ≥4 aneuploid chromosomes had the longest tPB1 and dMI (P < 0.0001), whereas eggs with one to three aneuploid chromosomes exhibited intermediate lengths of time (P < 0.0001). LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION We used Nocodazole in this study to disrupt the meiotic spindle and induce aneuploidy in mouse oocytes. Whether the association between morphokinetic variables of meiotic progression and the severity of aneuploidy occurs with other compounds that induce chromosome segregation errors remain to be investigated. In addition, unlike mouse oocytes, human IVM requires the presence of cumulus cells, which precludes visualization of morphokinetic variables of meiotic progression. Thus, our study may have limited direct clinical translatability. WIDER IMPLICATIONS OF THE FINDINGS We validated NGS in mouse eggs to detect aneuploidy at a chromosome-specific resolution which greatly improves the utility of the mouse model. With a tractable and validated model system for characterizing meiotic aneuploidy, investigations into the molecular mechanisms and factors which may influence aneuploidy can be further elaborated. Time-lapse analyses of morphokinetic variables of meiotic progression may be a useful non-invasive predictor of aneuploidy severity. STUDY FUNDING/COMPETING INTERESTS This work was supported by the Bill & Melinda Gates Foundation (INV-003385). Under the grant conditions of the Foundation, a Creative Commons Attribution 4.0 Generic License has already been assigned to the Author Accepted Manuscript version that might arise from this submission. The authors have no conflict of interest to disclose.
Collapse
Affiliation(s)
- Yiru Zhu
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Jeffrey Pea
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hoi Chang Lee
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Caroline E Kratka
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jia Xu
- Genomic Prediction Inc., North Brunswick Township, NJ, USA
| | - Diego Marin
- Genomic Prediction Inc., North Brunswick Township, NJ, USA
- Department of Genetics, Rutgers University, New Brunswick, NJ, USA
| | - Nathan R Treff
- Genomic Prediction Inc., North Brunswick Township, NJ, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
4
|
Rémillard-Labrosse G, Cohen S, Boucher É, Gagnon K, Vasilev F, Mihajlović AI, FitzHarris G. Oocyte and embryo culture under oil profoundly alters effective concentrations of small molecule inhibitors. Front Cell Dev Biol 2024; 12:1337937. [PMID: 38544820 PMCID: PMC10966923 DOI: 10.3389/fcell.2024.1337937] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/19/2024] [Indexed: 11/11/2024] Open
Abstract
Culture of oocytes and embryos in media under oil is a cornerstone of fertility treatment, and extensively employed in experimental investigation of early mammalian development. It has been noted anecdotally by some that certain small molecule inhibitors might lose activity in oil-covered culture systems, presumably by drug partitioning into the oil. Here we took a pseudo-pharmacological approach to appraise this formally using mouse oocytes and embryos. Using different culture dish designs with defined media:oil volume ratios, we show that the EC50 of the widely employed microtubule poison nocodazole shifts as a function of the media:oil ratio, such that nocodazole concentrations that prevent cell division in oil-free culture fail to in oil-covered media drops. Relatively subtle changes in culture dish design lead to measurable changes in EC50. This effect is not specific to one type of culture oil, and can be readily observed both in oocyte and embryo culture experiments. We subsequently applied a similar approach to a small panel of widely employed cell cycle-related inhibitors, finding that most lose activity in standard oil-covered oocyte/embryo culture systems. Our data suggest that loss of small molecule activity in oil-covered oocyte and embryo culture is a widespread phenomenon with potentially far-reaching implications for data reproducibility, and we recommend avoiding oil-covered culture for experiments employing inhibitors/drugs wherever possible.
Collapse
Affiliation(s)
| | - Sydney Cohen
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Éliane Boucher
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Kéryanne Gagnon
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Filip Vasilev
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Aleksandar I. Mihajlović
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Greg FitzHarris
- Centre de Recherche du Centre Hospitalier de I’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Obstetrics and Gynaecology, Université de Montréal, Montréal, QC, Canada
- Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
5
|
Horakova A, Konecna M, Anger M. Chromosome Division in Early Embryos-Is Everything under Control? And Is the Cell Size Important? Int J Mol Sci 2024; 25:2101. [PMID: 38396778 PMCID: PMC10889803 DOI: 10.3390/ijms25042101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Chromosome segregation in female germ cells and early embryonic blastomeres is known to be highly prone to errors. The resulting aneuploidy is therefore the most frequent cause of termination of early development and embryo loss in mammals. And in specific cases, when the aneuploidy is actually compatible with embryonic and fetal development, it leads to severe developmental disorders. The main surveillance mechanism, which is essential for the fidelity of chromosome segregation, is the Spindle Assembly Checkpoint (SAC). And although all eukaryotic cells carry genes required for SAC, it is not clear whether this pathway is active in all cell types, including blastomeres of early embryos. In this review, we will summarize and discuss the recent progress in our understanding of the mechanisms controlling chromosome segregation and how they might work in embryos and mammalian embryos in particular. Our conclusion from the current literature is that the early mammalian embryos show limited capabilities to react to chromosome segregation defects, which might, at least partially, explain the widespread problem of aneuploidy during the early development in mammals.
Collapse
Affiliation(s)
- Adela Horakova
- Department of Genetics and Reproductive Biotechnologies, Veterinary Research Institute, 621 00 Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Science, 277 21 Libechov, Czech Republic
- Faculty of Science, Masaryk University, 602 00 Brno, Czech Republic
| | - Marketa Konecna
- Department of Genetics and Reproductive Biotechnologies, Veterinary Research Institute, 621 00 Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Science, 277 21 Libechov, Czech Republic
- Faculty of Science, Masaryk University, 602 00 Brno, Czech Republic
| | - Martin Anger
- Department of Genetics and Reproductive Biotechnologies, Veterinary Research Institute, 621 00 Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Science, 277 21 Libechov, Czech Republic
| |
Collapse
|
6
|
Wang S, Wu X, Zhang M, Chang S, Guo Y, Song S, Dai S, Wu K, Zeng S. NET1 is a critical regulator of spindle assembly and actin dynamics in mouse oocytes. Reprod Biol Endocrinol 2024; 22:5. [PMID: 38169395 PMCID: PMC10759572 DOI: 10.1186/s12958-023-01177-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Neuroepithelial transforming gene 1 (NET1) is a RhoA subfamily guanine nucleotide exchange factor that governs a wide array of biological processes. However, its roles in meiotic oocyte remain unclear. We herein demonstrated that the NET1-HACE1-RAC1 pathway mediates meiotic defects in the progression of oocyte maturation. METHODS NET1 was reduced using a specific small interfering RNA in mouse oocytes. Spindle assembly, chromosomal alignment, the actin cap, and chromosomal spreads were visualized by immunostaining and analyzed under confocal microscopy. We also applied mass spectroscopy, and western blot analysis for this investigation. RESULTS Our results revealed that NET1 was localized to the nucleus at the GV stage, and that after GVBD, NET1 was localized to the cytoplasm and predominantly distributed around the chromosomes, commensurate with meiotic progression. NET1 resided in the cytoplasm and significantly accumulated on the spindle at the MI and MII stages. Mouse oocytes depleted of Net1 exhibited aberrant first polar body extrusion and asymmetric division defects. We also determined that Net1 depletion resulted in reduced RAC1 protein expression in mouse oocytes, and that NET1 protected RAC1 from degradation by HACE1, and it was essential for actin dynamics and meiotic spindle formation. Importantly, exogenous RAC1 expression in Net1-depleted oocytes significantly rescued these defects. CONCLUSIONS Our results suggest that NET1 exhibits multiple roles in spindle stability and actin dynamics during mouse oocyte meiosis.
Collapse
Affiliation(s)
- Shiwei Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xuan Wu
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Mengmeng Zhang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Siyu Chang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yajun Guo
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuang Song
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shizhen Dai
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Keliang Wu
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shenming Zeng
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| |
Collapse
|
7
|
Babayev E, Suebthawinkul C, Gokyer D, Parkes WS, Rivas F, Pavone ME, Hall AR, Pritchard MT, Duncan FE. Cumulus expansion is impaired with advanced reproductive age due to loss of matrix integrity and reduced hyaluronan. Aging Cell 2023; 22:e14004. [PMID: 37850336 PMCID: PMC10652338 DOI: 10.1111/acel.14004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023] Open
Abstract
Reproductive aging is associated with ovulatory defects. Age-related ovarian fibrosis partially contributes to this phenotype as short-term treatment with anti-fibrotic compounds improves ovulation in reproductively old mice. However, age-dependent changes that are intrinsic to the follicle may also be relevant. In this study, we used a mouse model to demonstrate that reproductive aging is associated with impaired cumulus expansion which is accompanied by altered morphokinetic behavior of cumulus cells as assessed by time-lapse microscopy. The extracellular matrix integrity of expanded cumulus-oocyte complexes is compromised with advanced age as evidenced by increased penetration of fluorescent nanoparticles in a particle exclusion assay and larger open spaces on scanning electron microscopy. Reduced hyaluronan (HA) levels, decreased expression of genes encoding HA-associated proteins (e.g., Ptx3 and Tnfaip6), and increased expression of inflammatory genes and matrix metalloproteinases underlie this loss of matrix integrity. Importantly, HA levels are decreased with age in follicular fluid of women, indicative of conserved reproductive aging mechanisms. These findings provide novel mechanistic insights into how defects in cumulus expansion contribute to age-related infertility and may serve as a target to extend reproductive longevity.
Collapse
Affiliation(s)
- Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Chanakarn Suebthawinkul
- Department of Obstetrics and Gynecology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Department of Obstetrics and Gynecology, Faculty of MedicineChulalongkorn UniversityBangkokThailand
| | - Dilan Gokyer
- Department of Obstetrics and Gynecology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Wendena S. Parkes
- Department of Pharmacology, Toxicology, & Therapeutics, Institute for Reproductive and Developmental SciencesUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Felipe Rivas
- Virginia Tech‐Wake Forest University School of Biomedical Engineering and SciencesWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Mary Ellen Pavone
- Department of Obstetrics and Gynecology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Adam R. Hall
- Virginia Tech‐Wake Forest University School of Biomedical Engineering and SciencesWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Michele T. Pritchard
- Department of Pharmacology, Toxicology, & Therapeutics, Institute for Reproductive and Developmental SciencesUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
8
|
Dipali SS, Suebthawinkul C, Burdette JE, Pavone ME, Duncan FE. Human follicular fluid elicits select dose- and age-dependent effects on mouse oocytes and cumulus-oocyte complexes in a heterologous in vitro maturation assay. Mol Hum Reprod 2023; 29:gaad039. [PMID: 37950499 PMCID: PMC10674105 DOI: 10.1093/molehr/gaad039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 10/11/2023] [Indexed: 11/12/2023] Open
Abstract
Follicular fluid (FF) is a primary microenvironment of the oocyte within an antral follicle. Although several studies have defined the composition of human FF in normal physiology and determined how it is altered in disease states, the direct impacts of human FF on the oocyte are not well understood. The difficulty of obtaining suitable numbers of human oocytes for research makes addressing such a question challenging. Therefore, we used a heterologous model in which we cultured mouse oocytes in human FF. To determine whether FF has dose-dependent effects on gamete quality, we performed in vitro maturation of denuded oocytes from reproductively young mice (6-12 weeks) in 10%, 50%, or 100% FF from participants of mid-reproductive age (32-36 years). FF impacted meiotic competence in a dose-dependent manner, with concentrations >10% inhibiting meiotic progression and resulting in spindle and chromosome alignment defects. We previously demonstrated that human FF acquires a fibro-inflammatory cytokine signature with age. Thus, to determine whether exposure to an aging FF microenvironment contributes to the age-dependent decrease in gamete quality, we matured denuded oocytes and cumulus-oocyte complexes (COCs) in FF from reproductively young (28-30 years) and old (40-42 years) participants. FF decreased meiotic progression of COCs, but not oocytes, from reproductively young and old (9-12 months) mice in an age-dependent manner. Moreover, FF had modest age-dependent impacts on mitochondrial aggregation in denuded oocytes and cumulus layer expansion dynamics in COCs, which may influence fertilization or early embryo development. Overall, these findings demonstrate that acute human FF exposure can impact select markers of mouse oocyte quality in both dose- and age-dependent manners.
Collapse
Affiliation(s)
- Shweta S Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Chanakarn Suebthawinkul
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Mary Ellen Pavone
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
9
|
Yang Q, Li H, Wang H, Chen W, Zeng X, Luo X, Xu J, Sun Y. Deletion of enzymes for de novo NAD + biosynthesis accelerated ovarian aging. Aging Cell 2023; 22:e13904. [PMID: 37332134 PMCID: PMC10497836 DOI: 10.1111/acel.13904] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/17/2023] [Accepted: 05/20/2023] [Indexed: 06/20/2023] Open
Abstract
Recent advances highlight the pivotal role of nicotinamide adenine dinucleotide (NAD+ ) in ovarian aging. However, the roles of de novo NAD+ biosynthesis on ovarian aging are still unknown. Here, we found that genetic ablation of Ido1 (indoleamine-2,3-dioxygenase 1) or Qprt (Quinolinate phosphoribosyl transferase), two critical genes in de novo NAD+ biosynthesis, resulted in decreased ovarian NAD+ levels in middle-aged mice, leading to subfertility, irregular estrous cycles, reduced ovarian reserve, and accelerated aging. Moreover, we observed impaired oocyte quality, characterized by increased reactive oxygen species and spindle anomalies, which ultimately led to reduced fertilization ability and impaired early embryonic development. A transcriptomic analysis of ovaries in both mutant and wild-type mice revealed alterations in gene expression related to mitochondrial metabolism. Our findings were further supported by the observation of impaired mitochondrial distribution and decreased mitochondrial membrane potential in the oocytes of knockout mice. Supplementation with nicotinamide riboside (NR), an NAD+ booster, in mutant mice increased ovarian reserve and improved oocyte quality. Our study highlights the importance of the NAD+ de novo pathway in middle-aged female fertility.
Collapse
Affiliation(s)
- Qingling Yang
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Hui Li
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Huan Wang
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Wenhui Chen
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xinxin Zeng
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xiaoyan Luo
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jianmin Xu
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yingpu Sun
- Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Reproduction and GeneticsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
10
|
Suebthawinkul C, Babayev E, Lee HC, Duncan FE. Morphokinetic parameters of mouse oocyte meiotic maturation and cumulus expansion are not affected by reproductive age or ploidy status. J Assist Reprod Genet 2023; 40:1197-1213. [PMID: 37012451 PMCID: PMC10239409 DOI: 10.1007/s10815-023-02779-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/17/2023] [Indexed: 04/05/2023] Open
Abstract
INTRODUCTION Morphokinetic analysis using a closed time-lapse monitoring system (EmbryoScope + ™) provides quantitative metrics of meiotic progression and cumulus expansion. The goal of this study was to use a physiologic aging mouse model, in which egg aneuploidy levels increase, to determine whether there are age-dependent differences in morphokinetic parameters of oocyte maturation. METHODS Denuded oocytes and intact cumulus-oocyte complexes (COCs) were isolated from reproductively young and old mice and in vitro matured in the EmbryoScope + ™. Morphokinetic parameters of meiotic progression and cumulus expansion were evaluated, compared between reproductively young and old mice, and correlated with egg ploidy status. RESULTS Oocytes from reproductively old mice were smaller than young counterparts in terms of GV area (446.42 ± 4.15 vs. 416.79 ± 5.24 µm2, p < 0.0001) and oocyte area (4195.71 ± 33.10 vs. 4081.62 ± 41.04 µm2, p < 0.05). In addition, the aneuploidy incidence was higher in eggs with advanced reproductive age (24-27% vs. 8-9%, p < 0.05). There were no differences in the morphokinetic parameters of oocyte maturation between oocytes from reproductively young and old mice with respect to time to germinal vesicle breakdown (GVBD) (1.03 ± 0.03 vs. 1.01 ± 0.04 h), polar body extrusion (PBE) (8.56 ± 0.11 vs. 8.52 ± 0.15 h), duration of meiosis I (7.58 ± 0.10 vs. 7.48 ± 0.11 h), and kinetics of cumulus expansion (0.093 ± 0.002 vs. 0.089 ± 0.003 µm/min). All morphokinetic parameters of oocyte maturation were similar between euploid and aneuploid eggs irrespective of age. CONCLUSION There is no association between age or ploidy and the morphokinetics of mouse oocyte in vitro maturation (IVM). Future studies are needed to evaluate whether there is an association between morphokinetic dynamics of mouse IVM and embryo developmental competence.
Collapse
Affiliation(s)
- Chanakarn Suebthawinkul
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hoi Chang Lee
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
11
|
Kincade JN, Hlavacek A, Akera T, Balboula AZ. Initial spindle positioning at the oocyte center protects against incorrect kinetochore-microtubule attachment and aneuploidy in mice. SCIENCE ADVANCES 2023; 9:eadd7397. [PMID: 36800430 PMCID: PMC9937575 DOI: 10.1126/sciadv.add7397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
Spindle positioning within the oocyte must be tightly regulated. In mice, the spindle is predominantly assembled at the oocyte center before its migration toward the cortex to achieve the highly asymmetric division, a characteristic of female meiosis. The significance of the initial central positioning of the spindle is largely unknown. We show that initial spindle positioning at the oocyte center is an insurance mechanism to avoid the premature exposure of the spindle to cortical CDC42 signaling, which perturbs proper kinetochore-microtubule attachments, leading to the formation of aneuploid gametes. These findings contribute to understanding why female gametes are notoriously associated with high rates of aneuploidy, the leading genetic cause of miscarriage and congenital abnormalities.
Collapse
Affiliation(s)
- Jessica N. Kincade
- Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Avery Hlavacek
- Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Takashi Akera
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ahmed Z. Balboula
- Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
12
|
Li C, Zhang H, Wu H, Li R, Wen D, Tang Y, Gao Z, Xu R, Lu S, Wei Q, Zhao X, Pan M, Ma B. Intermittent fasting reverses the declining quality of aged oocytes. Free Radic Biol Med 2023; 195:74-88. [PMID: 36581058 DOI: 10.1016/j.freeradbiomed.2022.12.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022]
Abstract
Decreased oocyte quality and compromised embryo development are particularly prevalent in older females, but the aging-related cellular processes and effective ameliorative approaches have not been fully characterized. Intermittent fasting (IF) can help improve health and extend lifespan; nevertheless, how it regulates reproductive aging and its mechanisms remain unclear. We used naturally aged mice to investigate the role of IF in reproduction and found that just one month of every-other-day fasting was sufficient to improve oocyte quality. IF not only increased antral follicle numbers and ovulation but also enhanced oocyte meiotic competence and embryonic development by improving both nuclear and cytoplasmic maturation in maternally aged oocytes. The beneficial effects of IF manifested as alleviation of spindle structure abnormalities and chromosome segregation errors and maintenance of the correct cytoplasmic organelle reorganization. Moreover, single-cell transcriptome analysis showed that the positive impact of IF on aged oocytes was mediated by restoration of the nicotinamide adenine dinucleotide (NAD+)/Sirt1-mediated antioxidant defense system, which eliminated excessive accumulated ROS to suppress DNA damage and apoptosis. Collectively, these findings suggest that IF is a feasible approach to protect oocytes against advanced maternal age-related oxidation damage and to improve the reproductive outcomes of aged females.
Collapse
Affiliation(s)
- Chan Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Hui Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Hao Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Ruoyu Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Dongxu Wen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Yaju Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Zhen Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Rui Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Sihai Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Qiang Wei
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China
| | - Xiaoe Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China.
| | - Menghao Pan
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China.
| | - Baohua Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Yangling, Shaanxi, People's Republic of China.
| |
Collapse
|
13
|
Takahashi N, Franciosi F, Daldello EM, Luong XG, Althoff P, Wang X, Conti M. CPEB1-dependent disruption of the mRNA translation program in oocytes during maternal aging. Nat Commun 2023; 14:416. [PMID: 36697412 PMCID: PMC9877008 DOI: 10.1038/s41467-023-35994-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/11/2023] [Indexed: 01/27/2023] Open
Abstract
The molecular causes of deteriorating oocyte quality during aging are poorly defined. Since oocyte developmental competence relies on post-transcriptional regulations, we tested whether defective mRNA translation contributes to this decline in quality. Disruption in ribosome loading on maternal transcripts is present in old oocytes. Using a candidate approach, we detect altered translation of 3'-UTR-reporters and altered poly(A) length of the endogenous mRNAs. mRNA polyadenylation depends on the cytoplasmic polyadenylation binding protein 1 (CPEB1). Cpeb1 mRNA translation and protein levels are decreased in old oocytes. This decrease causes de-repression of Ccnb1 translation in quiescent oocytes, premature CDK1 activation, and accelerated reentry into meiosis. De-repression of Ccnb1 is corrected by Cpeb1 mRNA injection in old oocytes. Oocyte-specific Cpeb1 haploinsufficiency in young oocytes recapitulates all the translation phenotypes of old oocytes. These findings demonstrate that a dysfunction in the oocyte translation program is associated with the decline in oocyte quality during aging.
Collapse
Affiliation(s)
- Nozomi Takahashi
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Federica Franciosi
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,Reproductive and Developmental Biology Lab, Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 20133, Milan, Italy
| | - Enrico Maria Daldello
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,Sorbonne Université, CNRS, Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Paris, France
| | - Xuan G Luong
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Peter Althoff
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Xiaotian Wang
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA.,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA
| | - Marco Conti
- Center for Reproductive Sciences, University of California, San Francisco, CA, 94143, USA. .,USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, 94143, USA. .,Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
14
|
Suebthawinkul C, Babayev E, Zhou LT, Lee HC, Duncan FE. Quantitative morphokinetic parameters identify novel dynamics of oocyte meiotic maturation and cumulus expansion†. Biol Reprod 2022; 107:1097-1112. [PMID: 35810327 PMCID: PMC9562117 DOI: 10.1093/biolre/ioac139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/30/2022] [Accepted: 06/30/2022] [Indexed: 12/05/2022] Open
Abstract
Meiotic maturation and cumulus expansion are essential for the generation of a developmentally competent gamete, and both processes can be recapitulated in vitro. We used a closed time-lapse incubator (EmbryoScope+™) to establish morphokinetic parameters of meiotic progression and cumulus expansion in mice and correlated these outcomes with egg ploidy. The average time to germinal vesicle breakdown (GVBD), time to first polar body extrusion (PBE), and duration of meiosis I were 0.91 ± 0.01, 8.82 ± 0.06, and 7.93 ± 0.06 h, respectively. The overall rate of cumulus layer expansion was 0.091 ± 0.002 μm/min, and the velocity of expansion peaked during the first 8 h of in vitro maturation (IVM) and then slowed. IVM of oocytes exposed to Nocodazole, a microtubule disrupting agent, and cumulus oocyte complexes (COCs) to 4-methylumbelliferone, a hyaluronan synthesis inhibitor, resulted in a dose-dependent perturbation of morphokinetics, thereby validating the system. The incidence of euploidy following IVM was >90% for both denuded oocytes and intact COCs. No differences were observed between euploid and aneuploid eggs with respect to time to GVBD (0.90 ± 0.22 vs. 0.97 ± 0.19 h), time to PBE (8.89 ± 0.98 vs. 9.10 ± 1.42 h), duration of meiosis I (8.01 ± 0.91 vs. 8.13 ± 1.38 h), and overall rate and kinetics of cumulus expansion (0.089 ± 0.02 vs 0.088 ± 0.03 μm/min) (P > 0.05). These morphokinetic parameters provide novel quantitative and non-invasive metrics for the evaluation of meiotic maturation and cumulus expansion and will enable screening compounds that modulate these processes.
Collapse
Affiliation(s)
- Chanakarn Suebthawinkul
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Luhan Tracy Zhou
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hoi Chang Lee
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
15
|
Chromosome Segregation in the Oocyte: What Goes Wrong during Aging. Int J Mol Sci 2022; 23:ijms23052880. [PMID: 35270022 PMCID: PMC8911062 DOI: 10.3390/ijms23052880] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/22/2022] [Accepted: 03/05/2022] [Indexed: 12/13/2022] Open
Abstract
Human female fertility and reproductive lifespan decrease significantly with age, resulting in an extended post-reproductive period. The central dogma in human female reproduction contains two important aspects. One is the pool of oocytes in the human ovary (the ovarian reserve; approximately 106 at birth), which diminishes throughout life until menopause around the age of 50 (approximately 103 oocytes) in women. The second is the quality of oocytes, including the correctness of meiotic divisions, among other factors. Notably, the increased rate of sub- and infertility, aneuploidy, miscarriages, and birth defects are associated with advanced maternal age, especially in women above 35 years of age. This postponement is also relevant for human evolution; decades ago, the female aging-related fertility drop was not as important as it is today because women were having their children at a younger age. Spindle assembly is crucial for chromosome segregation during each cell division and oocyte maturation, making it an important event for euploidy. Consequently, aberrations in this segregation process, especially during the first meiotic division in human eggs, can lead to implantation failure or spontaneous abortion. Today, human reproductive medicine is also facing a high prevalence of aneuploidy, even in young females. However, the shift in the reproductive phase of humans and the strong increase in errors make the problem much more dramatic at later stages of the female reproductive phase. Aneuploidy in human eggs could be the result of the non-disjunction of entire chromosomes or sister chromatids during oocyte meiosis, but partial or segmental aneuploidies are also relevant. In this review, we intend to describe the relevance of the spindle apparatus during oocyte maturation for proper chromosome segregation in the context of maternal aging and the female reproductive lifespan.
Collapse
|
16
|
Londoño-Vásquez D, Rodriguez-Lukey K, Behura SK, Balboula AZ. Microtubule organizing centers regulate spindle positioning in mouse oocytes. Dev Cell 2022; 57:197-211.e3. [PMID: 35030327 PMCID: PMC8792338 DOI: 10.1016/j.devcel.2021.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 10/18/2021] [Accepted: 12/10/2021] [Indexed: 01/26/2023]
Abstract
During female meiosis I (MI), spindle positioning must be tightly regulated to ensure the fidelity of the first asymmetric division and faithful chromosome segregation. Although the role of F-actin in regulating these critical processes has been studied extensively, little is known about whether microtubules (MTs) participate in regulating these processes. Using mouse oocytes as a model system, we characterize a subset of MT organizing centers that do not contribute directly to spindle assembly, termed mcMTOCs. Using laser ablation, STED super-resolution microscopy, and chemical manipulation, we show that mcMTOCs are required to regulate spindle positioning and faithful chromosome segregation during MI. We discuss how forces exerted by F-actin on the spindle are balanced by mcMTOC-nucleated MTs to anchor the spindle centrally and to regulate its timely migration. Our findings provide a model for asymmetric cell division, complementing the current F-actin-based models, and implicate mcMTOCs as a major player in regulating spindle positioning.
Collapse
Affiliation(s)
| | | | - Susanta K Behura
- Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
| | - Ahmed Z Balboula
- Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA; University of Cambridge, Department of Genetics, Downing Street, Cambridge, CB2 3EH, UK.
| |
Collapse
|
17
|
Blengini CS, Nguyen AL, Aboelenain M, Schindler K. Age-dependent integrity of the meiotic spindle assembly checkpoint in females requires Aurora kinase B. Aging Cell 2021; 20:e13489. [PMID: 34704342 PMCID: PMC8590096 DOI: 10.1111/acel.13489] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 08/09/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022] Open
Abstract
A hallmark of advanced maternal age is a significant increase in meiotic chromosome segregation errors, resulting in early miscarriages and congenital disorders. These errors most frequently occur during meiosis I (MI). The spindle assembly checkpoint (SAC) prevents chromosome segregation errors by arresting the cell cycle until proper chromosome alignment is achieved. Unlike in mitosis, the SAC in oocytes is desensitized, allowing chromosome segregation in the presence of improperly aligned chromosomes. Whether SAC integrity further deteriorates with advancing maternal age, and if this decline contributes to increased segregation errors remains a fundamental question. In somatic cells, activation of the SAC depends upon Aurora kinase B (AURKB), which functions to monitor kinetochore–microtubule attachments and recruit SAC regulator proteins. In mice, oocyte‐specific deletion of AURKB (Aurkb cKO) results in an increased production of aneuploid metaphase II‐arrested eggs and premature age‐related infertility. Here, we aimed to understand the cause of the short reproductive lifespan and hypothesized that SAC integrity was compromised. In comparing oocytes from young and sexually mature Aurkb cKO females, we found that SAC integrity becomes compromised rapidly with maternal age. We show that the increased desensitization of the SAC is driven by reduced expression of MAD2, ZW10 and Securin proteins, key contributors to the SAC response pathway. The reduced expression of these proteins is the result of altered protein homeostasis, likely caused by the accumulation of reactive oxygen species. Taken together, our results demonstrate a novel function for AURKB in preserving the female reproductive lifespan possibly by protecting oocytes from oxidative stress.
Collapse
Affiliation(s)
- Cecilia S. Blengini
- Department of Genetics; Rutgers The State University of New Jersey Piscataway NJ USA
- Human Genetics Institute of New Jersey Piscataway NJ USA
| | - Alexandra L. Nguyen
- Department of Genetics; Rutgers The State University of New Jersey Piscataway NJ USA
- Human Genetics Institute of New Jersey Piscataway NJ USA
| | - Mansour Aboelenain
- Department of Genetics; Rutgers The State University of New Jersey Piscataway NJ USA
- Human Genetics Institute of New Jersey Piscataway NJ USA
- Department of Theriogenology Faculty of Veterinary Medicine Mansoura University Mansoura Egypt
| | - Karen Schindler
- Department of Genetics; Rutgers The State University of New Jersey Piscataway NJ USA
- Human Genetics Institute of New Jersey Piscataway NJ USA
| |
Collapse
|
18
|
Balboula AZ, Schindler K, Kotani T, Kawahara M, Takahashi M. Vitrification-induced activation of lysosomal cathepsin B perturbs spindle assembly checkpoint function in mouse oocytes. Mol Hum Reprod 2021; 26:689-701. [PMID: 32634244 DOI: 10.1093/molehr/gaaa051] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/16/2020] [Indexed: 01/05/2023] Open
Abstract
As the age of child-bearing increases and correlates with infertility, cryopreservation of female gametes is becoming common-place in ART. However, the developmental competence of vitrified oocytes has remained low. The underlying mechanisms responsible for reduced oocyte quality post-vitrification are largely unknown. Mouse cumulus-oocyte complexes were vitrified using a cryoloop technique and a mixture of dimethylsulphoxide, ethylene glycol and trehalose as cryoprotectants. Fresh and vitrified/thawed oocytes were compared for chromosome alignment, spindle morphology, kinetochore-microtubule attachments, spindle assembly checkpoint (SAC) and aneuploidy. Although the majority of vitrified oocytes extruded the first polar body (PB), they had a significant increase of chromosome misalignment, abnormal spindle formation and aneuploidy at metaphase II. In contrast to controls, vitrified oocytes extruded the first PB in the presence of nocodazole and etoposide, which should induce metaphase I arrest in a SAC-dependent manner. The fluorescence intensity of mitotic arrest deficient 2 (MAD2), an essential SAC protein, at kinetochores was reduced in vitrified oocytes, indicating that the SAC is weakened after vitrification/thawing. Furthermore, we found that vitrification-associated stress disrupted lysosomal function and stimulated cathepsin B activity, with a subsequent activation of caspase 3. MAD2 localization and SAC function in vitrified oocytes were restored upon treatment with a cathepsin B or a caspase 3 inhibitor. This study was conducted using mouse oocytes, therefore confirming these results in human oocytes is a prerequisite before applying these findings in IVF clinics. Here, we uncovered underlying molecular pathways that contribute to an understanding of how vitrification compromises oocyte quality. Regulating these pathways will be a step toward improving oocyte quality post vitrification and potentially increasing the efficiency of the vitrification program.
Collapse
Affiliation(s)
- Ahmed Z Balboula
- Division of Animal Sciences, Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA.,Laboratory of Animal Breeding and Reproduction, Graduate school of Agriculture, Hokkaido University, Hokkaido 060-8589, Japan.,Department of Theriogenology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Karen Schindler
- Department of Genetics, Rutgers University, Piscataway, NJ 08854, USA
| | - Tomoya Kotani
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Manabu Kawahara
- Laboratory of Animal Breeding and Reproduction, Graduate school of Agriculture, Hokkaido University, Hokkaido 060-8589, Japan
| | - Masashi Takahashi
- Research Faculty of Agriculture, Hokkaido University, Hokkaido 060-8589, Japan.,Global Station for Food, Land and Water Resources, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido 060-0815, Japan
| |
Collapse
|
19
|
Distinct classes of lagging chromosome underpin age-related oocyte aneuploidy in mouse. Dev Cell 2021; 56:2273-2283.e3. [PMID: 34428397 DOI: 10.1016/j.devcel.2021.07.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/07/2021] [Accepted: 07/29/2021] [Indexed: 12/18/2022]
Abstract
Chromosome segregation errors that cause oocyte aneuploidy increase in frequency with maternal age and are considered a major contributing factor of age-related fertility decline in females. Lagging anaphase chromosomes are a common age-associated phenomenon in oocytes, but whether anaphase laggards actually missegregate and cause aneuploidy is unclear. Here, we show that lagging chromosomes in mouse oocytes comprise two mechanistically distinct classes of chromosome motion that we refer to as "class-I" and "class-II" laggards. We use imaging approaches and mechanistic interventions to dissociate the two classes and find that whereas class-II laggards are largely benign, class-I laggards frequently directly lead to aneuploidy. Most notably, a controlled prolongation of meiosis I specifically lessens class-I lagging to prevent aneuploidy. Our data thus reveal lagging chromosomes to be a cause of age-related aneuploidy in mouse oocytes and suggest that manipulating the cell cycle could increase the yield of useful oocytes in some contexts.
Collapse
|
20
|
Rogers HB, Zhou LT, Kusuhara A, Zaniker E, Shafaie S, Owen BC, Duncan FE, Woodruff TK. Dental resins used in 3D printing technologies release ovo-toxic leachates. CHEMOSPHERE 2021; 270:129003. [PMID: 33515896 PMCID: PMC7957323 DOI: 10.1016/j.chemosphere.2020.129003] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/31/2020] [Accepted: 11/15/2020] [Indexed: 06/02/2023]
Abstract
We recently engineered the first female reproductive tract on a chip (EVATAR), to enable sex-based ex vivo research. To increase the scalability and accessibility of EVATAR, we turned to 3D printing (3DP) technologies, selecting two biocompatible 3DP resins, Dental SG (DSG) and Dental LT (DLT) to generate 3DP microphysiologic platforms. Due to the known sensitivity of reproductive cells to leachable compounds, we first screened for toxicity of these biomaterials using an in vitro mammalian oocyte maturation assay. Culture of mouse oocytes in 3DP plates using conventionally treated DSG resin resulted in rapid oocyte degeneration. Oxygen plasma treatment of the surface of printed DSG resin prevented this degeneration, and the majority of the resulting oocytes progressed through meiosis in vitro. However, 57.0% ± 37.2% of the cells cultured in the DSG resin plates exhibited abnormal chromosome morphology compared to 19.4% ± 17.3% of controls cultured in polystyrene. All tested DLT resin conditions, including plasma treatment, resulted in complete and rapid oocyte degeneration. To identify the ovo-toxic component of DLT, we analyzed DLT leachate using mass spectroscopy. We identified Tinuvin 292, a commercial light stabilizer, as a major component of the DLT leachate, which resulted in a dose-dependent disruption of meiotic progression and increase in chromosomal abnormalities with oocyte exposure, showing significant ovo-toxicity in mammals. Severe reproductive toxicity induced by in vitro exposure to these 3D-printed resins highlights potential risks of deploying insufficiently characterized materials for biomedical applications and underscores the need for more rigorous evaluation and designation of biocompatible materials.
Collapse
Affiliation(s)
- Hunter B Rogers
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, 60611, USA
| | - Luhan T Zhou
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, 60611, USA
| | - Atsuko Kusuhara
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, 60611, USA
| | - Emily Zaniker
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, 60611, USA
| | - Saman Shafaie
- Integrated Molecular Structure Education and Research Center (IMSERC), Northwestern University, Evanston, IL, 60208, USA
| | - Benjamin C Owen
- Integrated Molecular Structure Education and Research Center (IMSERC), Northwestern University, Evanston, IL, 60208, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, 60611, USA.
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
21
|
Li A, Wang HX, Wang F, Fan LH, Zhao ZH, Han F, Li J, Lei WL, Zhou Q, Shi YP, Song CY, Schatten H, Sun QY, Guo XP. Nuclear and cytoplasmic quality of oocytes derived from serum-free culture of secondary follicles in vitro. J Cell Physiol 2021; 236:5352-5361. [PMID: 33586215 DOI: 10.1002/jcp.30232] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 12/28/2022]
Abstract
In vitro culture of follicles is a promising technology to generate large quantities of mature oocytes and it could offer a novel option of assisted reproductive technologies. Here we described a 2-dimensional follicular serum-free culture system with 3-dimensional effect that can make secondary follicles develop into antral follicles (78.52%), generating developmentally mature oocytes in vitro (66.45%). The oocytes in this serum-free system completed the first meiosis; spindle assembly and chromosome congression in most oocytes matured from follicular culture were normal. However, these oocytes showed significantly lower activation and embryonic development rates, and their ability to produce Ca2+ oscillations was also lower in response to parthenogenetic activation, after which a 2-cell embryonic developmental block occurred. Oocytes matured from follicular culture displayed increased abnormal mitochondrial distribution and increased reactive oxygen species levels when compared to in vivo matured oocytes. These data are important for understanding the reasons for reduced developmental potential of oocytes matured from follicular culture, and for further improving the cultivation system.
Collapse
Affiliation(s)
- Ang Li
- Faculty of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Province Reproductive Science Institute, Taiyuan, Shanxi, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Huai-Xiu Wang
- Shanxi Province Reproductive Science Institute, Taiyuan, Shanxi, China
| | | | - Li-Hua Fan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zheng-Hui Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Feng Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | - Wen-Long Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qian Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ya-Ping Shi
- Shanxi Province Reproductive Science Institute, Taiyuan, Shanxi, China
| | - Chun-Ying Song
- Shanxi Province Reproductive Science Institute, Taiyuan, Shanxi, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xing-Ping Guo
- Faculty of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Province Reproductive Science Institute, Taiyuan, Shanxi, China
| |
Collapse
|
22
|
Holton RA, Harris AM, Mukerji B, Singh T, Dia F, Berkowitz KM. CHTF18 ensures the quantity and quality of the ovarian reserve†. Biol Reprod 2020; 103:24-35. [PMID: 32219340 DOI: 10.1093/biolre/ioaa036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 10/29/2019] [Accepted: 03/24/2020] [Indexed: 11/13/2022] Open
Abstract
The number and quality of oocytes, as well as the decline in both of these parameters with age, determines reproductive potential in women. However, the underlying mechanisms of this diminution are incompletely understood. Previously, we identified novel roles for CHTF18 (Chromosome Transmission Fidelity Factor 18), a component of the conserved Replication Factor C-like complex, in male fertility and gametogenesis. Currently, we reveal crucial roles for CHTF18 in female meiosis and oocyte development. Chtf18-/- female mice are subfertile and have fewer offspring beginning at 6 months of age. Consistent with age-dependent subfertility, Chtf18-/- ovaries contain fewer follicles at all stages of folliculogenesis than wild type ovaries, but the decreases are more significant at 3 and 6 months of age. By 6 months of age, both primordial and growing ovarian follicle pools are markedly reduced to near depletion. Chromosomal synapsis in Chtf18-/- oocytes is complete, but meiotic recombination is impaired resulting in persistent DNA double-strand breaks, fewer crossovers, and early homolog disjunction during meiosis I. Consistent with poor oocyte quality, the majority of Chtf18-/- oocytes fail to progress to metaphase II following meiotic resumption and a significant percentage of those that do progress are aneuploid. Collectively, our findings indicate critical functions for CHTF18 in ensuring both the quantity and quality of the mammalian oocyte pool.
Collapse
Affiliation(s)
| | | | | | - Tanu Singh
- Department of Biochemistry and Molecular Biology
| | - Ferdusy Dia
- Department of Biochemistry and Molecular Biology
| | - Karen M Berkowitz
- Department of Biochemistry and Molecular Biology.,Department of Obstetrics and Gynecology, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
23
|
Li A, Wang F, Li L, Fan LH, Meng TG, Li QN, Wang Y, Yue W, Wang HX, Shi YP, Li HX, Schatten H, Sun QY, Guo XP. Mechanistic insights into the reduced developmental capacity of in vitro matured oocytes and importance of cumulus cells in oocyte quality determination. J Cell Physiol 2020; 235:9743-9751. [PMID: 32415704 DOI: 10.1002/jcp.29786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 04/12/2020] [Accepted: 04/29/2020] [Indexed: 12/17/2022]
Abstract
In vitro maturation of oocytes is a promising assisted reproductive technology (ART) for infertility treatment, although it is still not a routine technique for human ART due to reduced embryonic development. The aim of the present study was to clarify the possible reasons for reduced capacity of in vitro matured oocytes. Our results showed that the oocytes matured in vitro displayed increased abnormal mitochondrial distribution, reduced mitochondrial membrane potential, and increased reactive oxygen species levels when compared to in vivo matured oocytes. These results were not different in oocytes matured in vitro with or without cumulus cells. Notably, in vitro matured oocytes displayed increased mitochondrial DNA numbers probably due to functional compensation. In vitro matured oocytes showed significantly lower activation and embryonic development rates, and their ability to produce Ca2+ oscillations was much lower in response to parthenogenetic activation, especially in oocytes matured in vitro without cumulus cells with nearly half of them failing to produce calcium waves upon strontium chloride stimulation. These data are important for understanding the reasons for reduced developmental potential of in vitro matured oocytes and the importance of cumulus cells for oocyte quality.
Collapse
Affiliation(s)
- Ang Li
- Faculty of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China.,Shanxi Province Reproductive Science Institute, Taiyuan, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Feng Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Li Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Li-Hua Fan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Tie-Gang Meng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qian-Nan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yue Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wei Yue
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Huai-Xiu Wang
- Shanxi Province Reproductive Science Institute, Taiyuan, China
| | - Ya-Ping Shi
- Shanxi Province Reproductive Science Institute, Taiyuan, China
| | - Hong-Xia Li
- Shanxi Province Reproductive Science Institute, Taiyuan, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Qing-Yuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xing-Ping Guo
- Faculty of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China.,Shanxi Province Reproductive Science Institute, Taiyuan, China
| |
Collapse
|
24
|
Rémillard-Labrosse G, Dean NL, Allais A, Mihajlović AI, Jin SG, Son WY, Chung JT, Pansera M, Henderson S, Mahfoudh A, Steiner N, Agapitou K, Marangos P, Buckett W, Ligeti-Ruiter J, FitzHarris G. Human oocytes harboring damaged DNA can complete meiosis I. Fertil Steril 2020; 113:1080-1089.e2. [PMID: 32276763 DOI: 10.1016/j.fertnstert.2019.12.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 10/24/2022]
Abstract
OBJECTIVE To determine whether human oocytes possess a checkpoint to prevent completion of meiosis I when DNA is damaged. DESIGN DNA damage is considered a major threat to the establishment of healthy eggs and embryos. Recent studies found that mouse oocytes with damaged DNA can resume meiosis and undergo germinal vesicle breakdown (GVBD), but then arrest in metaphase of meiosis I in a process involving spindle assembly checkpoint (SAC) signaling. Such a mechanism could help prevent the generation of metaphase II (MII) eggs with damaged DNA. Here, we compared the impact of DNA-damaging agents with nondamaged control samples in mouse and human oocytes. SETTING University-affiliated clinic and research center. PATIENT(S) Patients undergoing ICSI cycles donated GV-stage oocytes after informed consent; 149 human oocytes were collected over 2 years (from 50 patients aged 27-44 years). INTERVENTIONS(S) Mice and human oocytes were treated with DNA-damaging drugs. MAIN OUTCOME MEASURE(S) Oocytes were monitored to evaluate GVBD and polar body extrusion (PBE), in addition to DNA damage assessment with the use of γH2AX antibodies and confocal microscopy. RESULT(S) Whereas DNA damage in mouse oocytes delays or prevents oocyte maturation, most human oocytes harboring experimentally induced DNA damage progress through meiosis I and subsequently form an MII egg, revealing the absence of a DNA damage-induced SAC response. Analysis of the resulting MII eggs revealed damaged DNA and chaotic spindle apparatus, despite the oocyte appearing morphologically normal. CONCLUSION(S) Our data indicate that experimentally induced DNA damage does not prevent PBE in human oocytes and can persist in morphologically normal looking MII eggs.
Collapse
Affiliation(s)
| | - Nicola L Dean
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Adélaïde Allais
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Aleksandar I Mihajlović
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Département d'Obstétrique-Gynécologie, Université de Montréal, Montreal, Quebec, Canada
| | - Shao Guang Jin
- Reproductive Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Weon-Young Son
- Reproductive Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jin-Tae Chung
- Reproductive Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Melissa Pansera
- Reproductive Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Sara Henderson
- Reproductive Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Alina Mahfoudh
- Reproductive Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Naama Steiner
- Reproductive Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Kristy Agapitou
- Department of Applications and Technology, University of Ioannina, Ioannina, Greece; Institute of Life Fertility Unit, IASO Maternity Hospital, Athens, Greece
| | - Petros Marangos
- Department of Applications and Technology, University of Ioannina, Ioannina, Greece; Department of Biomedical Research, Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology, Ioannina, Greece
| | - William Buckett
- Reproductive Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jacob Ligeti-Ruiter
- Reproductive Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Greg FitzHarris
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; Département d'Obstétrique-Gynécologie, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
25
|
Artificially decreasing cortical tension generates aneuploidy in mouse oocytes. Nat Commun 2020; 11:1649. [PMID: 32245998 PMCID: PMC7125192 DOI: 10.1038/s41467-020-15470-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 03/10/2020] [Indexed: 01/28/2023] Open
Abstract
Human and mouse oocytes’ developmental potential can be predicted by their mechanical properties. Their development into blastocysts requires a specific stiffness window. In this study, we combine live-cell and computational imaging, laser ablation, and biophysical measurements to investigate how deregulation of cortex tension in the oocyte contributes to early developmental failure. We focus on extra-soft cells, the most common defect in a natural population. Using two independent tools to artificially decrease cortical tension, we show that chromosome alignment is impaired in extra-soft mouse oocytes, despite normal spindle morphogenesis and dynamics, inducing aneuploidy. The main cause is a cytoplasmic increase in myosin-II activity that could sterically hinder chromosome capture. We describe here an original mode of generation of aneuploidies that could be very common in oocytes and could contribute to the high aneuploidy rate observed during female meiosis, a leading cause of infertility and congenital disorders. The developmental potential of human and murine oocytes is predicted by their mechanical properties. Here the authors show that artificial reduction of cortex tension produces aneuploid mouse oocytes and speculate that this may contribute to the high aneuploidy rate typical of female meiosis.
Collapse
|
26
|
Identifying the Translatome of Mouse NEBD-Stage Oocytes via SSP-Profiling; A Novel Polysome Fractionation Method. Int J Mol Sci 2020; 21:ijms21041254. [PMID: 32070012 PMCID: PMC7072993 DOI: 10.3390/ijms21041254] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/13/2022] Open
Abstract
Meiotic maturation of oocyte relies on pre-synthesised maternal mRNA, the translation of which is highly coordinated in space and time. Here, we provide a detailed polysome profiling protocol that demonstrates a combination of the sucrose gradient ultracentrifugation in small SW55Ti tubes with the qRT-PCR-based quantification of 18S and 28S rRNAs in fractionated polysome profile. This newly optimised method, named Scarce Sample Polysome Profiling (SSP-profiling), is suitable for both scarce and conventional sample sizes and is compatible with downstream RNA-seq to identify polysome associated transcripts. Utilising SSP-profiling we have assayed the translatome of mouse oocytes at the onset of nuclear envelope breakdown (NEBD)—a developmental point, the study of which is important for furthering our understanding of the molecular mechanisms leading to oocyte aneuploidy. Our analyses identified 1847 transcripts with moderate to strong polysome occupancy, including abundantly represented mRNAs encoding mitochondrial and ribosomal proteins, proteasomal components, glycolytic and amino acids synthetic enzymes, proteins involved in cytoskeleton organization plus RNA-binding and translation initiation factors. In addition to transcripts encoding known players of meiotic progression, we also identified several mRNAs encoding proteins of unknown function. Polysome profiles generated using SSP-profiling were more than comparable to those developed using existing conventional approaches, being demonstrably superior in their resolution, reproducibility, versatility, speed of derivation and downstream protocol applicability.
Collapse
|
27
|
Yi ZY, Meng TG, Ma XS, Li J, Zhang CH, Ouyang YC, Schatten H, Qiao J, Sun QY, Qian WP. CDC6 regulates both G2/M transition and metaphase-to-anaphase transition during the first meiosis of mouse oocytes. J Cell Physiol 2020; 235:5541-5554. [PMID: 31984513 DOI: 10.1002/jcp.29469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 01/06/2020] [Indexed: 12/30/2022]
Abstract
Cell division cycle protein, CDC6, is essential for the initiation of DNA replication. CDC6 was recently shown to inhibit the microtubule-organizing activity of the centrosome. Here, we show that CDC6 is localized to the spindle from pro-metaphase I (MI) to MII stages of oocytes, and it plays important roles at two critical steps of oocyte meiotic maturation. CDC6 depletion facilitated the G2/M transition (germinal vesicle breakdown [GVBD]) through regulation of Cdh1 and cyclin B1 expression and CDK1 (CDC2) phosphorylation in a GVBD-inhibiting culture system containing milrinone. Furthermore, GVBD was significantly decreased after knockdown of cyclin B1 in CDC6-depleted oocytes, indicating that the effect of CDC6 loss on GVBD stimulation was mediated, at least in part, by raising cyclin B1. Knockdown of CDC6 also caused abnormal localization of γ-tubulin, resulting in defective spindles, misaligned chromosomes, cyclin B1 accumulation, and spindle assembly checkpoint (SAC) activation, leading to significant pro-MI/MI arrest and PB1 extrusion failure. These phenotypes were also confirmed by time-lapse live cell imaging analysis. The results indicate that CDC6 is indispensable for maintaining G2 arrest of meiosis and functions in G2/M checkpoint regulation in mouse oocytes. Moreover, CDC6 is also a key player regulating meiotic spindle assembly and metaphase-to-anaphase transition in meiotic oocytes.
Collapse
Affiliation(s)
- Zi-Yun Yi
- The Reproductive Medicine Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Tie-Gang Meng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xue-Shan Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jian Li
- The Reproductive Medicine Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Chun-Hui Zhang
- The Reproductive Medicine Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Jie Qiao
- Reproductive Medical Center, Peking University Third Hospital, Beijing, China
| | - Qing-Yuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei-Ping Qian
- The Reproductive Medicine Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| |
Collapse
|
28
|
Dipali SS, Ferreira CR, Zhou LT, Pritchard MT, Duncan FE. Histologic analysis and lipid profiling reveal reproductive age-associated changes in peri-ovarian adipose tissue. Reprod Biol Endocrinol 2019; 17:46. [PMID: 31189477 PMCID: PMC6563378 DOI: 10.1186/s12958-019-0487-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Reproductive aging is a robust phenotype that occurs in all females and is characterized by a significant reduction in gamete quantity and quality, which can have negative consequences on both endocrine function and fertility. Age-associated differences in the oocyte, follicle, and ovary have been well-documented, but how the broader environment changes with age is less well understood. Fat is one of the largest organs in the body, and peri-gonadal adipose tissue surrounds the rodent ovary and comprises a local ovarian environment. The goal of this study was to characterize how peri-ovarian adipose tissue changes with advanced reproductive age. METHODS We isolated peri-gonadal adipose tissue from two cohorts of CB6F1 mice: reproductively young (6-12 weeks) and reproductively old (14-17 months). A comparative histological analysis was performed to evaluate adipocyte architecture. We then extracted lipids from the tissue and performed multiple reaction monitoring (MRM)-profiling, a mass spectrometry-based method of metabolite profiling, to compare the lipid profiles of peri-gonadal adipose tissue in these age cohorts. RESULTS We found that advanced reproductive age was associated with adipocyte hypertrophy and a corresponding decrease in the number of adipocytes per area. Of the 10 lipid classes examined, triacylglycerols (TAGs) had significantly different profiles between young and old cohorts, despite quantitative analysis revealing a decrease in the total amount of TAGs per weight of peri-gonadal adipose tissue with age. CONCLUSIONS These findings pinpoint age-associated physiological changes in peri-gonadal adipose tissue with respect to adipocyte morphology and lipid profiles and lay the foundation for future studies to examine how these alterations may influence both adipocyte and ovarian function.
Collapse
Affiliation(s)
- Shweta S Dipali
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 7-117, Chicago, IL, 60611, USA
| | - Christina R Ferreira
- Center for Analytical Instrumentation Development, Department of Chemistry, Purdue University, West Lafayette, IN, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | - Luhan T Zhou
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 7-117, Chicago, IL, 60611, USA
| | - Michele T Pritchard
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 7-117, Chicago, IL, 60611, USA.
| |
Collapse
|
29
|
Namgoong S, Kim NH. Meiotic spindle formation in mammalian oocytes: implications for human infertility. Biol Reprod 2019; 98:153-161. [PMID: 29342242 DOI: 10.1093/biolre/iox145] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022] Open
Abstract
In the final stage of oogenesis, mammalian oocytes generate a meiotic spindle and undergo chromosome segregation to yield an egg that is ready for fertilization. Herein, we describe the recent advances in understanding the mechanisms controlling formation of the meiotic spindle in metaphase I (MI) and metaphase II (MII) in mammalian oocytes, and focus on the differences between mouse and human oocytes. Unlike mitotic cells, mammalian oocytes lack typical centrosomes that consist of two centrioles and the surrounding pericentriolar matrix proteins, which serve as microtubule-organizing centers (MTOCs) in most somatic cells. Instead, oocytes rely on different mechanisms for the formation of microtubules in MI spindles. Two different mechanisms have been described for MI spindle formation in mammalian oocytes. Chromosome-mediated microtubule formation, including RAN-mediated spindle formation and chromosomal passenger complex-mediated spindle elongation, controls the growth of microtubules from chromatin, while acentriolar MTOC-mediated microtubule formation contributes to spindle formation. Mouse oocytes utilize both chromatin- and MTOC-mediated pathways for microtubule formation. The existence of both pathways may provide a fail-safe mechanism to ensure high fidelity of chromosome segregation during meiosis. Unlike mouse oocytes, human oocytes considered unsuitable for clinical in vitro fertilization procedures, lack MTOCs; this may explain why meiosis in human oocytes is often error-prone. Understanding the mechanisms of MI/MII spindle formation, spindle assembly checkpoint, and chromosome segregation, in mammalian oocytes, will provide valuable insights into the molecular mechanisms of human infertility.
Collapse
Affiliation(s)
| | - Nam-Hyung Kim
- Department of Animal Science, Chungbuk National University, Cheong-Ju, Chungbuk, Republic of Korea
| |
Collapse
|
30
|
Krisher RL. Maternal age affects oocyte developmental potential at both ends of the age spectrum. Reprod Fertil Dev 2019; 31:1-9. [PMID: 32188537 DOI: 10.1071/rd18340] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Maternal age has a significant effect on oocyte developmental competence. Overall, evidence suggests that oocytes from both prepubertal females and reproductively aged females are inherently less competent. Reduced oocyte quality in both age groups is problematic for human medicine and agriculture. Some of the cellular mechanisms implicated in poor oocyte quality associated with maternal age are mitochondrial function and location, reduction of oxygen radicals, balance of metabolic pathways, regulation of maternal mRNAs and appropriate communication between the oocyte and cumulus cells. However, additional knowledge must be gained about the deficiencies present in prepubertal and reproductively aged oocytes that result in poor developmental potential before significant improvement can be achieved. This review discusses the evidence currently available regarding oocyte quality at both ends of the maternal age spectrum, what we know, or hypothesise, about the mechanisms involved and current thoughts regarding potential treatment for improvement.
Collapse
Affiliation(s)
- Rebecca L Krisher
- Colorado Center for Reproductive Medicine, 10290 RidgeGate Circle, Lone Tree, CO 80124, USA. Email
| |
Collapse
|
31
|
Pasquariello R, Ermisch AF, Silva E, McCormick S, Logsdon D, Barfield JP, Schoolcraft WB, Krisher RL. Alterations in oocyte mitochondrial number and function are related to spindle defects and occur with maternal aging in mice and humans†. Biol Reprod 2018; 100:971-981. [DOI: 10.1093/biolre/ioy248] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/29/2018] [Accepted: 11/19/2018] [Indexed: 01/09/2023] Open
Affiliation(s)
- Rolando Pasquariello
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado, USA
- Colorado State University, College of Veterinary Medicine and Biomedical Sciences, Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Fort Collins, Colorado, USA
| | - Alison F Ermisch
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado, USA
| | - Elena Silva
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado, USA
| | - Sue McCormick
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado, USA
| | - Deirdre Logsdon
- Colorado Center for Reproductive Medicine, Lone Tree, Colorado, USA
| | - Jennifer P Barfield
- Colorado State University, College of Veterinary Medicine and Biomedical Sciences, Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Fort Collins, Colorado, USA
| | | | | |
Collapse
|
32
|
Brown A, Geiger H. Chromosome integrity checkpoints in stem and progenitor cells: transitions upon differentiation, pathogenesis, and aging. Cell Mol Life Sci 2018; 75:3771-3779. [PMID: 30066086 PMCID: PMC6154040 DOI: 10.1007/s00018-018-2891-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 07/22/2018] [Accepted: 07/25/2018] [Indexed: 01/30/2023]
Abstract
Loss of chromosome integrity is a major contributor to cancer. Checkpoints within the cell division cycle that facilitate the accuracy and outcome of chromosome segregation are thus critical pathways for preserving chromosome integrity and preventing chromosomal instability. The spindle assembly checkpoint, the decatenation checkpoint and the post-mitotic tetraploidy checkpoint ensure the appropriate establishment of the spindle apparatus, block mitotic entry upon entanglement of chromosomes or prevent further progression of post-mitotic cells that display massive spindle defects. Most of our knowledge on these mechanisms originates from studies conducted in yeast, cancer cell lines and differentiated cells. Considering that in many instances cancer derives from transformed stem and progenitor cells, our knowledge on these checkpoints in these cells just started to emerge. With this review, we provide a general overview of the current knowledge of these checkpoints in embryonic as well as in adult stem and progenitor cells with a focus on the hematopoietic system and outline common mis-regulations of their function associated with cancer and leukemia. Most cancers are aging-associated diseases. We will thus also discuss changes in the function and outcome of these checkpoints upon aging of stem and progenitor cells.
Collapse
Affiliation(s)
- Andreas Brown
- Institute of Molecular Medicine, Ulm University, Life Science Building N27, James Franck-Ring/Meyerhofstrasse, 89081, Ulm, Germany
| | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Life Science Building N27, James Franck-Ring/Meyerhofstrasse, 89081, Ulm, Germany.
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
| |
Collapse
|
33
|
Increased Expression of Maturation Promoting Factor Components Speeds Up Meiosis in Oocytes from Aged Females. Int J Mol Sci 2018; 19:ijms19092841. [PMID: 30235877 PMCID: PMC6164426 DOI: 10.3390/ijms19092841] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/12/2018] [Accepted: 09/17/2018] [Indexed: 12/24/2022] Open
Abstract
The rate of chromosome segregation errors that emerge during meiosis I in the mammalian female germ line are known to increase with maternal age; however, little is known about the underlying molecular mechanism. The objective of this study was to analyze meiotic progression of mouse oocytes in relation to maternal age. Using the mouse as a model system, we analyzed the timing of nuclear envelope breakdown and the morphology of the nuclear lamina of oocytes obtained from young (2 months old) and aged females (12 months old). Oocytes obtained from older females display a significantly faster progression through meiosis I compared to the ones obtained from younger females. Furthermore, in oocytes from aged females, lamin A/C structures exhibit rapid phosphorylation and dissociation. Additionally, we also found an increased abundance of MPF components and increased translation of factors controlling translational activity in the oocytes of aged females. In conclusion, the elevated MPF activity observed in aged female oocytes affects precocious meiotic processes that can multifactorially contribute to chromosomal errors in meiosis I.
Collapse
|
34
|
Franciosi F, Goudet G, Tessaro I, Papillier P, Dalbies-Tran R, Reigner F, Deleuze S, Douet C, Miclea I, Lodde V, Luciano AM. In vitro maturation affects chromosome segregation, spindle morphology and acetylation of lysine 16 on histone H4 in horse oocytes. Reprod Fertil Dev 2018; 29:721-730. [PMID: 26651296 DOI: 10.1071/rd15350] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/12/2015] [Indexed: 12/14/2022] Open
Abstract
Implantation failure and genetic developmental disabilities in mammals are caused by errors in chromosome segregation originating mainly in the oocyte during meiosis I. Some conditions, like maternal ageing or in vitro maturation (IVM), increase the incidence of oocyte aneuploidy. Here oocytes from adult mares were used to investigate oocyte maturation in a monovulatory species. Experiments were conducted to compare: (1) the incidence of aneuploidy, (2) the morphology of the spindle, (3) the acetylation of lysine 16 on histone H4 (H4K16) and (4) the relative amount of histone acetyltransferase 1 (HAT1), K(lysine) acetyltransferase 8 (KAT8, also known as MYST1), histone deacetylase 1 (HDAC1) and NAD-dependent protein deacetylase sirtuin 1 (SIRT1) mRNA in metaphase II stage oocytes that were in vitro matured or collected from peri-ovulatory follicles. The frequency of aneuploidy and anomalies in spindle morphology was increased following IVM, along with a decrease in H4K16 acetylation that was in agreement with our previous observations. However, differences in the amount of the transcripts investigated were not detected. These results suggest that the degradation of transcripts encoding for histone deacetylases and acetyltransferases is not involved in the changes of H4K16 acetylation observed following IVM, while translational or post-translational mechanisms might have a role. Our study also suggests that epigenetic instabilities introduced by IVM may affect the oocyte and embryo genetic stability.
Collapse
Affiliation(s)
- Federica Franciosi
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Science and Food Safety, University of Milan, via Celoria, 10, Milan, 20133, Italy
| | - Ghylene Goudet
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, F-37380, France
| | - Irene Tessaro
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Science and Food Safety, University of Milan, via Celoria, 10, Milan, 20133, Italy
| | - Pascal Papillier
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, F-37380, France
| | - Rozenn Dalbies-Tran
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, F-37380, France
| | | | - Stefan Deleuze
- Université de Liège, Clinique des Animaux de Compagnie et des Équidés, Place du 20 Août 7, Liège, 4000, Belgium
| | - Cecile Douet
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, F-37380, France
| | - Ileana Miclea
- University of Agricultural Sciences and Veterinary Medicine, Calea M?n?tur 3-5, Cluj-Napoca 400372, Romania
| | - Valentina Lodde
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Science and Food Safety, University of Milan, via Celoria, 10, Milan, 20133, Italy
| | - Alberto M Luciano
- Reproductive and Developmental Biology Laboratory, Department of Health, Animal Science and Food Safety, University of Milan, via Celoria, 10, Milan, 20133, Italy
| |
Collapse
|
35
|
The Post-anaphase SUMO Pathway Ensures the Maintenance of Centromeric Cohesion through Meiosis I-II Transition in Mammalian Oocytes. Curr Biol 2018; 28:1661-1669.e4. [PMID: 29754905 DOI: 10.1016/j.cub.2018.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/23/2018] [Accepted: 04/05/2018] [Indexed: 12/27/2022]
Abstract
The production of haploid gametes requires the maintenance of centromeric cohesion between sister chromatids through the transition between two successive meiotic divisions, meiosis I and meiosis II. One mechanism for the cohesion maintenance is shugoshin-dependent protection of centromeric cohesin at anaphase I onset [1-3]. However, how centromeric cohesion is maintained during late anaphase I and telophase I, when centromeric shugoshin is undetectable [1-3], remains largely unexplored. Here we show that the centromeric small ubiquitin-related modifier (SUMO) pathway is critical for the maintenance of centromeric cohesion during post-anaphase-I periods in mouse oocytes. SUMO2/3 and E3 ligase PIAS are enriched near centromeres during late anaphase I and telophase I. Specific perturbation of the centromeric SUMO pathway results in precocious loss of centromeric cohesin at telophase I, although shugoshin-dependent centromeric protection at anaphase I onset remains largely intact. Prevention of the SUMO perturbation during post-anaphase-I periods restores the maintenance of centromeric cohesion through the meiosis I-II transition. Thus, the post-anaphase-I centromeric SUMO pathway ensures continuous maintenance of centromeric cohesion through the meiosis I-II transition.
Collapse
|
36
|
Greaney J, Wei Z, Homer H. Regulation of chromosome segregation in oocytes and the cellular basis for female meiotic errors. Hum Reprod Update 2017; 24:135-161. [PMID: 29244163 DOI: 10.1093/humupd/dmx035] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 09/12/2017] [Accepted: 11/26/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Meiotic chromosome segregation in human oocytes is notoriously error-prone, especially with ageing. Such errors markedly reduce the reproductive chances of increasing numbers of women embarking on pregnancy later in life. However, understanding the basis for these errors is hampered by limited access to human oocytes. OBJECTIVE AND RATIONALE Important new discoveries have arisen from molecular analyses of human female recombination and aneuploidy along with high-resolution analyses of human oocyte maturation and mouse models. Here, we review these findings to provide a contemporary picture of the key players choreographing chromosome segregation in mammalian oocytes and the cellular basis for errors. SEARCH METHODS A search of PubMed was conducted using keywords including meiosis, oocytes, recombination, cohesion, cohesin complex, chromosome segregation, kinetochores, spindle, aneuploidy, meiotic cell cycle, spindle assembly checkpoint, anaphase-promoting complex, DNA damage, telomeres, mitochondria, female ageing and female fertility. We extracted papers focusing on mouse and human oocytes that best aligned with the themes of this review and that reported transformative and novel discoveries. OUTCOMES Meiosis incorporates two sequential rounds of chromosome segregation executed by a spindle whose component microtubules bind chromosomes via kinetochores. Cohesion mediated by the cohesin complex holds chromosomes together and should be resolved at the appropriate time, in a specific step-wise manner and in conjunction with meiotically programmed kinetochore behaviour. In women, the stage is set for meiotic error even before birth when female-specific crossover maturation inefficiency leads to the formation of at-risk recombination patterns. In adult life, multiple co-conspiring factors interact with at-risk crossovers to increase the likelihood of mis-segregation. Available evidence support that these factors include, but are not limited to, cohesion deterioration, uncoordinated sister kinetochore behaviour, erroneous microtubule attachments, spindle instability and structural chromosomal defects that impact centromeres and telomeres. Data from mice indicate that cohesin and centromere-specific histones are long-lived proteins in oocytes. Since these proteins are pivotal for chromosome segregation, but lack any obvious renewal pathway, their deterioration with age provides an appealing explanation for at least some of the problems in older oocytes. WIDER IMPLICATIONS Research in the mouse model has identified a number of candidate genes and pathways that are important for chromosome segregation in this species. However, many of these have not yet been investigated in human oocytes so it is uncertain at this stage to what extent they apply to women. The challenge for the future involves applying emerging knowledge of female meiotic molecular regulation towards improving clinical fertility management.
Collapse
Affiliation(s)
- Jessica Greaney
- Christopher Chen Oocyte Biology Research Laboratory, Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital Campus, Herston QLD 4029, Australia
| | - Zhe Wei
- Christopher Chen Oocyte Biology Research Laboratory, Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital Campus, Herston QLD 4029, Australia
| | - Hayden Homer
- Christopher Chen Oocyte Biology Research Laboratory, Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane & Women's Hospital Campus, Herston QLD 4029, Australia
| |
Collapse
|
37
|
Li X, Liu X, Gao M, Han L, Qiu D, Wang H, Xiong B, Sun SC, Liu H, Gu L. HDAC3 promotes meiotic apparatus assembly in mouse oocytes by modulating tubulin acetylation. Development 2017; 144:3789-3797. [PMID: 28935703 DOI: 10.1242/dev.153353] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/30/2017] [Indexed: 12/17/2022]
Abstract
Histone deacetylases (HDACs) have been shown to deacetylate numerous cellular substrates that govern a wide array of biological processes. HDAC3, a member of the Class I HDACs, is a highly conserved and ubiquitously expressed protein. However, its roles in meiotic oocytes are not known. In the present study, we find that mouse oocytes depleted of HDAC3 are unable to completely progress through meiosis, and are blocked at metaphase I. These HDAC3 knockdown oocytes show spindle/chromosome organization failure, with severely impaired kinetochore-microtubule attachments. Consistent with this, the level of BubR1, a central component of the spindle assembly checkpoint, at kinetochores is dramatically increased in metaphase oocytes following HDAC3 depletion. Knockdown and overexpression experiments reveal that HDAC3 modulates the acetylation status of α-tubulin in mouse oocytes. Importantly, the deacetylation mimetic mutant tubulin-K40R can partly rescue the defective phenotypes of HDAC3 knockdown oocytes. Our data support a model whereby HDAC3, through deacetylating tubulin, promotes microtubule stability and the establishment of kinetochore-microtubule interaction, consequently ensuring proper spindle morphology, accurate chromosome movement and orderly meiotic progression during oocyte maturation.
Collapse
Affiliation(s)
- Xiaoyan Li
- College of Animal Science & Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Xiaohui Liu
- College of Animal Science & Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Min Gao
- College of Animal Science & Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Longsen Han
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 211166 Nanjing, China
| | - Danhong Qiu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 211166 Nanjing, China
| | - Haichao Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 211166 Nanjing, China
| | - Bo Xiong
- College of Animal Science & Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Shao-Chen Sun
- College of Animal Science & Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Honglin Liu
- College of Animal Science & Technology, Nanjing Agricultural University, 210095 Nanjing, China
| | - Ling Gu
- College of Animal Science & Technology, Nanjing Agricultural University, 210095 Nanjing, China
| |
Collapse
|
38
|
Quartuccio SM, Dipali SS, Schindler K. Haspin inhibition reveals functional differences of interchromatid axis-localized AURKB and AURKC. Mol Biol Cell 2017; 28:2233-2240. [PMID: 28659416 PMCID: PMC5555651 DOI: 10.1091/mbc.e16-12-0850] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 06/16/2017] [Accepted: 06/20/2017] [Indexed: 01/11/2023] Open
Abstract
Use of mouse oocytes that only express Aurora kinase B as the catalytic subunit of the chromosomal passenger complex (CPC) provides evidence indicating differential capacities of AURKB– and AURKC–CPC complexes at a distinct localization. Aneuploidy is the leading genetic abnormality contributing to infertility, and chromosome segregation errors are common during female mammalian meiosis I (MI). Previous results indicate that haspin kinase regulates resumption of meiosis from prophase arrest, chromosome condensation, and kinetochore–microtubule attachments during early prometaphase of MI. Here we report that haspin inhibition in late prometaphase I causes acceleration of MI, bypass of the spindle assembly checkpoint (SAC), and loss of interchromatid axis–localized Aurora kinase C. Meiotic cells contain a second chromosomal passenger complex (CPC) population, with Aurora kinase B (AURKB) bound to INCENP. Haspin inhibition in oocytes from Aurkc−/− mice, where AURKB is the sole CPC kinase, does not alter MI completion timing, and no change in localization of the SAC protein, MAD2, is observed. These data suggest that AURKB on the interchromatid axis is not needed for SAC activation and illustrate a key difference between the functional capacities of the two AURK homologues.
Collapse
Affiliation(s)
- Suzanne M Quartuccio
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | - Shweta S Dipali
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| |
Collapse
|
39
|
Maternal age-dependent APC/C-mediated decrease in securin causes premature sister chromatid separation in meiosis II. Nat Commun 2017; 8:15346. [PMID: 28516917 PMCID: PMC5454377 DOI: 10.1038/ncomms15346] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 03/21/2017] [Indexed: 12/27/2022] Open
Abstract
Sister chromatid attachment during meiosis II (MII) is maintained by securin-mediated inhibition of separase. In maternal ageing, oocytes show increased inter-sister kinetochore distance and premature sister chromatid separation (PSCS), suggesting aberrant separase activity. Here, we find that MII oocytes from aged mice have less securin than oocytes from young mice and that this reduction is mediated by increased destruction by the anaphase promoting complex/cyclosome (APC/C) during meiosis I (MI) exit. Inhibition of the spindle assembly checkpoint (SAC) kinase, Mps1, during MI exit in young oocytes replicates this phenotype. Further, over-expression of securin or Mps1 protects against the age-related increase in inter-sister kinetochore distance and PSCS. These findings show that maternal ageing compromises the oocyte SAC–APC/C axis leading to a decrease in securin that ultimately causes sister chromatid cohesion loss. Manipulating this axis and/or increasing securin may provide novel therapeutic approaches to alleviating the risk of oocyte aneuploidy in maternal ageing. Sister chromatid cohesion during meiosis II (MII), maintained by securin-mediated inhibition of separase, is reduced in aged mouse oocytes. Here the authors show that, in MII oocytes, securin levels are reduced by increased destruction by the anaphase promoting complex/cyclosome.
Collapse
|
40
|
Franciosi F, Tessaro I, Dalbies-Tran R, Douet C, Reigner F, Deleuze S, Papillier P, Miclea I, Lodde V, Luciano AM, Goudet G. Analysis of Chromosome Segregation, Histone Acetylation, and Spindle Morphology in Horse Oocytes. J Vis Exp 2017. [PMID: 28518085 DOI: 10.3791/55242] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The field of assisted reproduction has been developed to treat infertility in women, companion animals, and endangered species. In the horse, assisted reproduction also allows for the production of embryos from high performers without interrupting their sports career and contributes to an increase in the number of foals from mares of high genetic value. The present manuscript describes the procedures used for collecting immature and mature oocytes from horse ovaries using ovum pick-up (OPU). These oocytes were then used to investigate the incidence of aneuploidy by adapting a protocol previously developed in mice. Specifically, the chromosomes and the centromeres of metaphase II (MII) oocytes were fluorescently labeled and counted on sequential focal plans after confocal laser microscope scanning. This analysis revealed a higher incidence in the aneuploidy rate when immature oocytes were collected from the follicles and matured in vitro compared to in vivo. Immunostaining for tubulin and the acetylated form of histone four at specific lysine residues also revealed differences in the morphology of the meiotic spindle and in the global pattern of histone acetylation. Finally, the expression of mRNAs coding for histone deacetylases (HDACs) and acetyl-transferases (HATs) was investigated by reverse transcription and quantitative-PCR (q-PCR). No differences in the relative expression of transcripts were observed between in vitro and in vivo matured oocytes. In agreement with a general silencing of the transcriptional activity during oocyte maturation, the analysis of the total transcript amount can only reveal mRNA stability or degradation. Therefore, these findings indicate that other translational and post-translational regulations might be affected. Overall, the present study describes an experimental approach to morphologically and biochemically characterize the horse oocyte, a cell type that is extremely challenging to study due to low sample availability. However, it can expand our knowledge on the reproductive biology and infertility in monovulatory species.
Collapse
Affiliation(s)
- Federica Franciosi
- Department of Health, Animal Science and Food Safety, University of Milan;
| | - Irene Tessaro
- Department of Health, Animal Science and Food Safety, University of Milan; IRCCS. Istituto Ortopedico Galeazzi
| | | | | | | | - Stefan Deleuze
- Clinique des Animaux de Compagnie et des Équidés, Université de Liège
| | | | - Ileana Miclea
- University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Valentina Lodde
- Department of Health, Animal Science and Food Safety, University of Milan
| | - Alberto M Luciano
- Department of Health, Animal Science and Food Safety, University of Milan
| | | |
Collapse
|
41
|
Zhang K, Smith GW. Maternal control of early embryogenesis in mammals. Reprod Fertil Dev 2017; 27:880-96. [PMID: 25695370 DOI: 10.1071/rd14441] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/10/2015] [Indexed: 12/11/2022] Open
Abstract
Oocyte quality is a critical factor limiting the efficiency of assisted reproductive technologies (ART) and pregnancy success in farm animals and humans. ART success is diminished with increased maternal age, suggesting a close link between poor oocyte quality and ovarian aging. However, the regulation of oocyte quality remains poorly understood. Oocyte quality is functionally linked to ART success because the maternal-to-embryonic transition (MET) is dependent on stored maternal factors, which are accumulated in oocytes during oocyte development and growth. The MET consists of critical developmental processes, including maternal RNA depletion and embryonic genome activation. In recent years, key maternal proteins encoded by maternal-effect genes have been determined, primarily using genetically modified mouse models. These proteins are implicated in various aspects of early embryonic development, including maternal mRNA degradation, epigenetic reprogramming, signal transduction, protein translation and initiation of embryonic genome activation. Species differences exist in the number of cell divisions encompassing the MET and maternal-effect genes controlling this developmental window. Perturbations of maternal control, some of which are associated with ovarian aging, result in decreased oocyte quality.
Collapse
Affiliation(s)
- Kun Zhang
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI 48824, USA
| | - George W Smith
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
42
|
Danadova J, Matijescukova N, Danylevska AMG, Anger M. Increased frequency of chromosome congression defects and aneuploidy in mouse oocytes cultured at lower temperature. Reprod Fertil Dev 2017; 29:968-974. [DOI: 10.1071/rd15306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 01/19/2016] [Indexed: 11/23/2022] Open
Abstract
Optimal culture conditions are essential for successful IVM of mammalian oocytes and for their further development into an embryo. In the present study we used live cell imaging microscopy to assess the effects of suboptimal culture temperature on various aspects of IVM, including duration of meiosis I, dynamics of polar body extrusion, chromosome congression, anaphase-promoting complex/cyclosome (APC/C) activation and aneuploidy. The data showed that even a small deviation from the optimal incubation temperature causes marked changes in the duration and synchronicity of meiosis, APC/C activity and the frequency of chromosome congression and segregation errors. In vitro manipulation and maturation of germ cells is widely used in both human and animal artificial reproduction techniques. Mammalian oocytes are naturally prone to chromosomal segregation errors, which are responsible for severe mental and developmental disorders. The data presented herein demonstrate that exposure of mouse oocytes to suboptimal temperature during manipulation and maturation could further increase the frequency of chromosome segregation defects in these cells.
Collapse
|
43
|
Treff NR, Krisher RL, Tao X, Garnsey H, Bohrer C, Silva E, Landis J, Taylor D, Scott RT, Woodruff TK, Duncan FE. Next Generation Sequencing-Based Comprehensive Chromosome Screening in Mouse Polar Bodies, Oocytes, and Embryos. Biol Reprod 2016; 94:76. [PMID: 26911429 DOI: 10.1095/biolreprod.115.135483] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/11/2016] [Indexed: 11/01/2022] Open
Abstract
Advanced reproductive age is unequivocally associated with increased aneuploidy in human oocytes, which contributes to infertility, miscarriages, and birth defects. The frequency of meiotic chromosome segregation errors in oocytes derived from reproductively aged mice appears to be similar to that observed in humans, but a limitation of this important model system is our inability to accurately identify chromosome-specific aneuploidy. Here we report the validation and application of a new low-pass whole-genome sequencing approach to comprehensively screen chromosome aneuploidy in individual mouse oocytes and blastocysts. First, we validated this approach by using single mouse embryonic fibroblasts engineered to have stable trisomy 16. We further validated this method by identifying reciprocal chromosome segregation errors in the products of meiosis I (gamete and polar body) in oocytes from reproductively aged mice. Finally, we applied this technology to investigate the incidence of aneuploidy in blastocysts derived from in vitro- and in vivo-matured oocytes in both young and reproductively aged mice. Using this next generation sequencing approach, we quantitatively assessed meiotic and mitotic segregation errors at the single chromosome level, distinguished between errors due to premature separation of sister chromatids and classical nondisjunction of homologous chromosomes, and quantified mitochondrial DNA (mtDNA) segregation in individual cells. This whole-genome sequencing technique, therefore, greatly improves the utility of the mouse model system for the study of aneuploidy and is a powerful quantitative tool with which to examine the molecular underpinnings of mammalian gamete and early embryo chromosome segregation in the context of reproductive aging and beyond.
Collapse
Affiliation(s)
- Nathan R Treff
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Reproductive Medicine Associates of New Jersey, Basking Ridge, New Jersey Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | | | - Xin Tao
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Reproductive Medicine Associates of New Jersey, Basking Ridge, New Jersey
| | - Heather Garnsey
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Reproductive Medicine Associates of New Jersey, Basking Ridge, New Jersey
| | - Chelsea Bohrer
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Reproductive Medicine Associates of New Jersey, Basking Ridge, New Jersey
| | - Elena Silva
- National Foundation for Fertility Research, Lone Tree, Colorado
| | - Jessica Landis
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Reproductive Medicine Associates of New Jersey, Basking Ridge, New Jersey Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Deanne Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Reproductive Medicine Associates of New Jersey, Basking Ridge, New Jersey Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Richard T Scott
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Reproductive Medicine Associates of New Jersey, Basking Ridge, New Jersey Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Francesca E Duncan
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
44
|
DNA damage-induced metaphase I arrest is mediated by the spindle assembly checkpoint and maternal age. Nat Commun 2015; 6:8706. [PMID: 26522734 PMCID: PMC4667640 DOI: 10.1038/ncomms9706] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/22/2015] [Indexed: 12/21/2022] Open
Abstract
In mammalian oocytes DNA damage can cause chromosomal abnormalities that potentially lead to infertility and developmental disorders. However, there is little known about the response of oocytes to DNA damage. Here we find that oocytes with DNA damage arrest at metaphase of the first meiosis (MI). The MI arrest is induced by the spindle assembly checkpoint (SAC) because inhibiting the SAC overrides the DNA damage-induced MI arrest. Furthermore, this MI checkpoint is compromised in oocytes from aged mice. These data lead us to propose that the SAC is a major gatekeeper preventing the progression of oocytes harbouring DNA damage. The SAC therefore acts to integrate protection against both aneuploidy and DNA damage by preventing production of abnormal mature oocytes and subsequent embryos. Finally, we suggest escaping this DNA damage checkpoint in maternal ageing may be one of the causes of increased chromosome anomalies in oocytes and embryos from older mothers.
Collapse
|
45
|
MacLennan M, Crichton JH, Playfoot CJ, Adams IR. Oocyte development, meiosis and aneuploidy. Semin Cell Dev Biol 2015; 45:68-76. [PMID: 26454098 PMCID: PMC4828587 DOI: 10.1016/j.semcdb.2015.10.005] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/14/2015] [Accepted: 10/05/2015] [Indexed: 01/15/2023]
Abstract
Meiosis is one of the defining events in gametogenesis. Male and female germ cells both undergo one round of meiotic cell division during their development in order to reduce the ploidy of the gametes, and thereby maintain the ploidy of the species after fertilisation. However, there are some aspects of meiosis in the female germline, such as the prolonged arrest in dictyate, that appear to predispose oocytes to missegregate their chromosomes and transmit aneuploidies to the next generation. These maternally-derived aneuploidies are particularly problematic in humans where they are major contributors to miscarriage, age-related infertility, and the high incidence of Down's syndrome in human conceptions. This review will discuss how events that occur in foetal oocyte development and during the oocytes' prolonged dictyate arrest can influence meiotic chromosome segregation and the incidence of aneuploidy in adult oocytes.
Collapse
Affiliation(s)
- Marie MacLennan
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| | - James H Crichton
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| | - Christopher J Playfoot
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| | - Ian R Adams
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| |
Collapse
|
46
|
Liu XJ. Targeting oocyte maturation to improve fertility in older women. Cell Tissue Res 2015; 363:57-68. [PMID: 26329301 DOI: 10.1007/s00441-015-2264-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/08/2015] [Indexed: 11/28/2022]
Abstract
Reproductive aging is an increasingly pressing problem facing women in modern society, due to delay in child bearing. According to Statistics Canada, 52% of all Canadian births in 2011 were by women aged 30 years and older, up from 24% in 1981 ( http://www.statcan.gc.ca/pub/91-209-x/2013001/article/11784-eng.htm ). Women older than 35 years of age experience significantly increased risks of infertility, miscarriage and congenital birth defects, mostly due to poor quality of the eggs. Increasingly sophisticated, and often invasive, assisted reproductive technologies (ARTs) have helped millions of women to achieve reproductive success. However, by and large, ARTs do not address the fundamental issue of reproductive aging in women: age-related decline in egg quality. More importantly, ARTs are not, and will never be, the main solution for the general population. Here, I attempt to review the scientific literature on age-related egg quality decline, based mostly on studies in mice and in humans. Emphasis is given to the brief period of time called oocyte maturation, which occurs just prior to ovulation. The rationale for this emphasis is that oocyte maturation represents a critical window where unfavorable ovarian conditions in older females contribute significantly to the decline of egg quality, and that science-based intervention during oocyte maturation represents the best chance of improving egg quality in older women. Finally, I summarize our own work in recent years on peri-ovulatory putrescine supplementation as a possible remedy for reproductive aging.
Collapse
Affiliation(s)
- X Johné Liu
- Ottawa Hospital Research Institute, The Ottawa Hospital - General Campus, 501 Smyth Road, Box 511, Ottawa, Ontario, K1H 8L6, Canada. .,Department of Obstetrics and Gynecology and Department of Biochemistry, Microbiology and Immunology (BMI), University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
47
|
Touati SA, Wassmann K. How oocytes try to get it right: spindle checkpoint control in meiosis. Chromosoma 2015; 125:321-35. [PMID: 26255654 DOI: 10.1007/s00412-015-0536-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/09/2015] [Accepted: 07/20/2015] [Indexed: 11/27/2022]
Abstract
The generation of a viable, diploid organism depends on the formation of haploid gametes, oocytes, and spermatocytes, with the correct number of chromosomes. Halving the genome requires the execution of two consecutive specialized cell divisions named meiosis I and II. Unfortunately, and in contrast to male meiosis, chromosome segregation in oocytes is error prone, with human oocytes being extraordinarily "meiotically challenged". Aneuploid oocytes, that are with the wrong number of chromosomes, give rise to aneuploid embryos when fertilized. In humans, most aneuploidies are lethal and result in spontaneous abortions. However, some trisomies survive to birth or even adulthood, such as the well-known trisomy 21, which gives rise to Down syndrome (Nagaoka et al. in Nat Rev Genet 13:493-504, 2012). A staggering 20-25 % of oocytes ready to be fertilized are aneuploid in humans. If this were not bad enough, there is an additional increase in meiotic missegregations as women get closer to menopause. A woman above 40 has a risk of more than 30 % of getting pregnant with a trisomic child. Worse still, in industrialized western societies, child birth is delayed, with women getting their first child later in life than ever. This trend has led to an increase of trisomic pregnancies by 70 % in the last 30 years (Nagaoka et al. in Nat Rev Genet 13:493-504, 2012; Schmidt et al. in Hum Reprod Update 18:29-43, 2012). To understand why errors occur so frequently during the meiotic divisions in oocytes, we review here the molecular mechanisms at works to control chromosome segregation during meiosis. An important mitotic control mechanism, namely the spindle assembly checkpoint or SAC, has been adapted to the special requirements of the meiotic divisions, and this review will focus on our current knowledge of SAC control in mammalian oocytes. Knowledge on how chromosome segregation is controlled in mammalian oocytes may help to identify risk factors important for questions related to human reproductive health.
Collapse
Affiliation(s)
- Sandra A Touati
- Institut de Biologie Paris Seine (IBPS), UMR7622, Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, France.,Chromosome Segregation Laboratory, Lincoln's Inn Fields Laboratory, The Francis Crick Institute, London, UK
| | - Katja Wassmann
- Institut de Biologie Paris Seine (IBPS), UMR7622, Sorbonne Universités, UPMC Univ Paris 06, Paris, France. .,CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, France.
| |
Collapse
|
48
|
Xiao S, Duncan FE, Bai L, Nguyen CT, Shea LD, Woodruff TK. Size-specific follicle selection improves mouse oocyte reproductive outcomes. Reproduction 2015; 150:183-92. [PMID: 26116002 DOI: 10.1530/rep-15-0175] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/18/2015] [Indexed: 12/14/2022]
Abstract
Encapsulated in vitro follicle growth (eIVFG) has great potential to provide an additional fertility preservation option for young women and girls with cancer or other reproductive health threatening diseases. Currently, follicles are cultured for a defined period of time and analyzed as a cohort. However, follicle growth is not synchronous, and culturing follicles for insufficient or excessive times can result in compromised gamete quality. Our objective is to determine whether the selection of follicles based on size, rather than absolute culture time, better predict follicle maturity and oocyte quality. Multilayer secondary mouse follicles were isolated and encapsulated in 0.25% alginate. Follicles were cultured individually either for defined time periods or up to specific follicle diameter ranges, at which point several reproductive endpoints were analyzed. The metaphase II (MII) percentage after oocyte maturation on day 6 was the highest (85%) when follicles were cultured for specific days. However, if follicles were cultured to a terminal diameter of 300-350 μm irrespective of absolute time in culture, 93% of the oocytes reached MII. More than 90% of MII oocytes matured from follicles with diameters of 300-350 μm showed normal spindle morphology and chromosome alignment, 85% of oocytes showed two pronuclei after IVF, 81% developed into the two-cell embryo stage and 38% developed to the blastocyst stage, all significantly higher than the percentages in the other follicle size groups. Our study demonstrates that size-specific follicle selection can be used as a non-invasive marker to identify high-quality oocytes and improve reproductive outcomes during eIVFG.
Collapse
Affiliation(s)
- Shuo Xiao
- Department of Obstetrics and GynecologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USACenter for Reproductive ScienceNorthwestern University, Evanston, Illinois 60208, USADepartment of Anatomy and Cell BiologyUniversity of Kansas Medical Center, Kansas City, Kansas 66160, USAMaster of Biotechnology ProgramNorthwestern University, Evanston, Illinois 60628, USADepartment of Biomedical EngineeringCollege of Engineering and Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA Department of Obstetrics and GynecologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USACenter for Reproductive ScienceNorthwestern University, Evanston, Illinois 60208, USADepartment of Anatomy and Cell BiologyUniversity of Kansas Medical Center, Kansas City, Kansas 66160, USAMaster of Biotechnology ProgramNorthwestern University, Evanston, Illinois 60628, USADepartment of Biomedical EngineeringCollege of Engineering and Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Francesca E Duncan
- Department of Obstetrics and GynecologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USACenter for Reproductive ScienceNorthwestern University, Evanston, Illinois 60208, USADepartment of Anatomy and Cell BiologyUniversity of Kansas Medical Center, Kansas City, Kansas 66160, USAMaster of Biotechnology ProgramNorthwestern University, Evanston, Illinois 60628, USADepartment of Biomedical EngineeringCollege of Engineering and Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Lu Bai
- Department of Obstetrics and GynecologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USACenter for Reproductive ScienceNorthwestern University, Evanston, Illinois 60208, USADepartment of Anatomy and Cell BiologyUniversity of Kansas Medical Center, Kansas City, Kansas 66160, USAMaster of Biotechnology ProgramNorthwestern University, Evanston, Illinois 60628, USADepartment of Biomedical EngineeringCollege of Engineering and Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA Department of Obstetrics and GynecologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USACenter for Reproductive ScienceNorthwestern University, Evanston, Illinois 60208, USADepartment of Anatomy and Cell BiologyUniversity of Kansas Medical Center, Kansas City, Kansas 66160, USAMaster of Biotechnology ProgramNorthwestern University, Evanston, Illinois 60628, USADepartment of Biomedical EngineeringCollege of Engineering and Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Catherine T Nguyen
- Department of Obstetrics and GynecologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USACenter for Reproductive ScienceNorthwestern University, Evanston, Illinois 60208, USADepartment of Anatomy and Cell BiologyUniversity of Kansas Medical Center, Kansas City, Kansas 66160, USAMaster of Biotechnology ProgramNorthwestern University, Evanston, Illinois 60628, USADepartment of Biomedical EngineeringCollege of Engineering and Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA Department of Obstetrics and GynecologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USACenter for Reproductive ScienceNorthwestern University, Evanston, Illinois 60208, USADepartment of Anatomy and Cell BiologyUniversity of Kansas Medical Center, Kansas City, Kansas 66160, USAMaster of Biotechnology ProgramNorthwestern University, Evanston, Illinois 60628, USADepartment of Biomedical EngineeringCollege of Engineering and Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Lonnie D Shea
- Department of Obstetrics and GynecologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USACenter for Reproductive ScienceNorthwestern University, Evanston, Illinois 60208, USADepartment of Anatomy and Cell BiologyUniversity of Kansas Medical Center, Kansas City, Kansas 66160, USAMaster of Biotechnology ProgramNorthwestern University, Evanston, Illinois 60628, USADepartment of Biomedical EngineeringCollege of Engineering and Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Teresa K Woodruff
- Department of Obstetrics and GynecologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USACenter for Reproductive ScienceNorthwestern University, Evanston, Illinois 60208, USADepartment of Anatomy and Cell BiologyUniversity of Kansas Medical Center, Kansas City, Kansas 66160, USAMaster of Biotechnology ProgramNorthwestern University, Evanston, Illinois 60628, USADepartment of Biomedical EngineeringCollege of Engineering and Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA Department of Obstetrics and GynecologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USACenter for Reproductive ScienceNorthwestern University, Evanston, Illinois 60208, USADepartment of Anatomy and Cell BiologyUniversity of Kansas Medical Center, Kansas City, Kansas 66160, USAMaster of Biotechnology ProgramNorthwestern University, Evanston, Illinois 60628, USADepartment of Biomedical EngineeringCollege of Engineering and Medical School, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
49
|
Touati SA, Buffin E, Cladière D, Hached K, Rachez C, van Deursen JM, Wassmann K. Mouse oocytes depend on BubR1 for proper chromosome segregation but not for prophase I arrest. Nat Commun 2015; 6:6946. [PMID: 25897860 DOI: 10.1038/ncomms7946] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 03/16/2015] [Indexed: 12/19/2022] Open
Abstract
Mammalian female meiosis is error prone, with rates of meiotic chromosome missegregations strongly increasing towards the end of the reproductive lifespan. A strong reduction of BubR1 has been observed in oocytes of women approaching menopause and in ovaries of aged mice, which led to the hypothesis that a gradual decline of BubR1 contributes to age-related aneuploidization. Here we employ a conditional knockout approach in mouse oocytes to dissect the meiotic roles of BubR1. We show that BubR1 is required for diverse meiotic functions, including persistent spindle assembly checkpoint activity, timing of meiosis I and the establishment of robust kinetochore-microtubule attachments in a meiosis-specific manner, but not prophase I arrest. These data reveal that BubR1 plays a multifaceted role in chromosome segregation during the first meiotic division and suggest that age-related decline of BubR1 is a key determinant of the formation of aneuploid oocytes as women approach menopause.
Collapse
Affiliation(s)
- Sandra A Touati
- Sorbonne Universités, UPMC Université Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, 75005 Paris, France.,CNRS, IBPS, UMR7622 Developmental Biology Lab, 75005 Paris, France
| | - Eulalie Buffin
- Sorbonne Universités, UPMC Université Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, 75005 Paris, France.,CNRS, IBPS, UMR7622 Developmental Biology Lab, 75005 Paris, France
| | - Damien Cladière
- Sorbonne Universités, UPMC Université Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, 75005 Paris, France.,CNRS, IBPS, UMR7622 Developmental Biology Lab, 75005 Paris, France
| | - Khaled Hached
- Sorbonne Universités, UPMC Université Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, 75005 Paris, France.,CNRS, IBPS, UMR7622 Developmental Biology Lab, 75005 Paris, France
| | - Christophe Rachez
- Departement de Biologie du Développement et Cellules Souches, CNRS URA2578, Unité de Régulation Epigénétique, Institut Pasteur, 75015 Paris, France
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine and Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | - Katja Wassmann
- Sorbonne Universités, UPMC Université Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, 75005 Paris, France.,CNRS, IBPS, UMR7622 Developmental Biology Lab, 75005 Paris, France
| |
Collapse
|
50
|
Herbert M, Kalleas D, Cooney D, Lamb M, Lister L. Meiosis and maternal aging: insights from aneuploid oocytes and trisomy births. Cold Spring Harb Perspect Biol 2015; 7:a017970. [PMID: 25833844 DOI: 10.1101/cshperspect.a017970] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In most organisms, genome haploidization requires reciprocal DNA exchanges (crossovers) between replicated parental homologs to form bivalent chromosomes. These are resolved to their four constituent chromatids during two meiotic divisions. In female mammals, bivalents are formed during fetal life and remain intact until shortly before ovulation. Extending this period beyond ∼35 years greatly increases the risk of aneuploidy in human oocytes, resulting in a dramatic increase in infertility, miscarriage, and birth defects, most notably trisomy 21. Bivalent chromosomes are stabilized by cohesion between sister chromatids, which is mediated by the cohesin complex. In mouse oocytes, cohesin becomes depleted from chromosomes during female aging. Consistent with this, premature loss of centromeric cohesion is a major source of aneuploidy in oocytes from older women. Here, we propose a mechanistic framework to reconcile data from genetic studies on human trisomy and oocytes with recent advances in our understanding of the molecular mechanisms of chromosome segregation during meiosis in model organisms.
Collapse
Affiliation(s)
- Mary Herbert
- Newcastle Fertility Centre, Centre for Life, Times Square, Newcastle upon Tyne NE1 4EP, United Kingdom Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Dimitrios Kalleas
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Daniel Cooney
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Mahdi Lamb
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Lisa Lister
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| |
Collapse
|